1
|
Lee S, Lee B, Kwon SH, Park J, Kim SH. MCC in the spotlight: Its dual role in signal regulation and oncogenesis. Cell Signal 2025; 131:111756. [PMID: 40118128 DOI: 10.1016/j.cellsig.2025.111756] [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: 01/24/2025] [Revised: 03/13/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
The mutated in colorectal cancer (MCC) gene is closely associated with the onset and progression of colorectal cancer. MCC plays a critical role in regulating the cell cycle and various signaling pathways and is recognized to inhibit cancer cell proliferation via the β-catenin signaling pathway. β-catenin is a key component of the WNT signaling pathway that influences cell growth, differentiation, survival, and migration, thereby positioning MCC as an important tumor suppressor. Notably, MCC has also been implicated in other cancer types, including lung, liver, and brain cancers. However, the precise mechanisms by which MCC functions in these malignancies remain inadequately understood. Comprehensive investigations into the interactions among MCC, various signaling pathways, and metabolic processes are essential for uncovering the molecular mechanisms of cancer and the pathological features characteristic of different cancer stages. This review presents the structural characteristics of MCC and its cell growth regulation mechanisms and functional roles within tissues, with the aims of enhancing our understanding of the role of MCC in cancer biology and highlighting potential therapeutic strategies targeting this gene.
Collapse
Affiliation(s)
- Soohyeon Lee
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 35015, South Korea; Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, South Korea
| | - Beomwoo Lee
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 35015, South Korea; Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, South Korea
| | - So Hee Kwon
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 21983, South Korea.
| | - Jongsun Park
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 35015, South Korea; Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, South Korea; Biomedical Research Institute, Chungnam National University Hospital, Daejeon 35015, Republic of Korea.
| | - Seon-Hwan Kim
- Biomedical Research Institute, Chungnam National University Hospital, Daejeon 35015, Republic of Korea; Department of Neurosurgery, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, South Korea.
| |
Collapse
|
2
|
Jomova K, Alomar SY, Valko R, Liska J, Nepovimova E, Kuca K, Valko M. Flavonoids and their role in oxidative stress, inflammation, and human diseases. Chem Biol Interact 2025; 413:111489. [PMID: 40147618 DOI: 10.1016/j.cbi.2025.111489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 02/23/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Oxidative stress and chronic inflammation are important drivers in the pathogenesis and progression of many chronic diseases, such as cancers of the breast, kidney, lung, and others, autoimmune diseases (rheumatoid arthritis), cardiovascular diseases (hypertension, atherosclerosis, arrhythmia), neurodegenerative diseases (Alzheimer's disease, Parkinson's disease, Huntington's disease), mental disorders (depression, schizophrenia, bipolar disorder), gastrointestinal disorders (inflammatory bowel disease, colorectal cancer), and other disorders. With the increasing demand for less toxic and more tolerable therapies, flavonoids have the potential to effectively modulate the responsiveness to conventional therapy and radiotherapy. Flavonoids are polyphenolic compounds found in fruits, vegetables, grains, and plant-derived beverages. Six of the twelve structurally different flavonoid subgroups are of dietary significance and include anthocyanidins (e.g. pelargonidin, cyanidin), flavan-3-ols (e.g. epicatechin, epigallocatechin), flavonols (e.g. quercetin, kaempferol), flavones (e.g. luteolin, baicalein), flavanones (e.g. hesperetin, naringenin), and isoflavones (daidzein, genistein). The health benefits of flavonoids are related to their structural characteristics, such as the number and position of hydroxyl groups and the presence of C2C3 double bonds, which predetermine their ability to chelate metal ions, terminate ROS (e.g. hydroxyl radicals formed by the Fenton reaction), and interact with biological targets to trigger a biological response. Based on these structural characteristics, flavonoids can exert both antioxidant or prooxidant properties, modulate the activity of ROS-scavenging enzymes and the expression and activation of proinflammatory cytokines (e.g., interleukin-1beta (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α)), induce apoptosis and autophagy, and target key signaling pathways, such as the nuclear factor erythroid 2-related factor 2 (Nrf2) and Bcl-2 family of proteins. This review aims to briefly discuss the mutually interconnected aspects of oxidative and inflammatory mechanisms, such as lipid peroxidation, protein oxidation, DNA damage, and the mechanism and resolution of inflammation. The major part of this article discusses the role of flavonoids in alleviating oxidative stress and inflammation, two common components of many human diseases. The results of epidemiological studies on flavonoids are also presented.
Collapse
Affiliation(s)
- Klaudia Jomova
- Department of Chemistry, Faculty of Natural Sciences, Constantine the Philosopher University in Nitra, Nitra, 949 74, Slovakia
| | - Suliman Y Alomar
- Zoology Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Richard Valko
- Zoology Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Jan Liska
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, 811 08, Bratislava, Slovakia
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Sciences, University of Hradec Kralove, 50003, Hradec Kralove, Czech Republic; Center of Advanced Innovation Technologies, VSB-Technical University of Ostrava, Ostrava-Poruba, 708 00, Czech Republic
| | - Kamil Kuca
- Center of Advanced Innovation Technologies, VSB-Technical University of Ostrava, Ostrava-Poruba, 708 00, Czech Republic; Biomedical Research Center, University Hospital Hradec Kralove, 5005, Hradec Kralove, Czech Republic
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, 812 37, Bratislava, Slovakia.
| |
Collapse
|
3
|
Yue S, Gong L, Tan Y, Zhang X, Liao F. IsoalloLCA-intervened regulatory T cell exosomes alleviate inflammatory bowel disease by inhibiting NF-κB-associated inflammation in intestinal epithelial cells. Int Immunopharmacol 2025; 154:114501. [PMID: 40174336 DOI: 10.1016/j.intimp.2025.114501] [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: 11/17/2024] [Revised: 02/11/2025] [Accepted: 03/14/2025] [Indexed: 04/04/2025]
Abstract
Regulatory T cells (Tregs) are the principal immune cells that exert anti-inflammatory effects within the organism. Their exosomes exhibit therapeutic efficacy across a broad spectrum of diseases owing to their high stability, low immunogenicity, and substantial penetration capacity. Recent research have indicated that isoallolithocholic acid (isoalloLCA), a metabolite associated with bile acid metabolism, may enhance Treg activity by upregulating forkhead box protein3 (Foxp3) expression. Hence, metabolite-based strategies for reinforcing Tregs may offer novel intervention options for treating related diseases. In this study, tumor necrosis factor (TNF)-α and dextran sulfate sodium (DSS) were employed to establish cellular and animal models of inflammatory bowel disease (IBD), further evaluating the therapeutic efficacy of isoalloLCA-intervened regulatory T cell exosomes (isoalloLCA-Exo) within this model. Our findings demonstrated that isoalloLCA-Exo effectively inhibit colitis progression in a murine model, as indicated by reduced inflammation, decreased apoptosis of intestinal epithelial cells, and improved intestinal barrier function. Furthermore, in vitro analyses elucidated the molecular mechanisms underlying the anti-inflammatory effects of isoalloLCA-Exo, revealing that the intervention effectively reversed TNF-α-induced inflammation and apoptosis in intestinal epithelial cells by modulating the NF-κB pathway. In conclusion, isoalloLCA-Exo can decelerate inflammatory bowel disease progression and suppress inflammatory response in intestinal epithelial cells by inhibiting NF-κB pathway. Notably, isoalloLCA-Exo exhibit superior efficacy to the traditional drug mesalazine and conventional treg exosome(NC-Exo). These findings have significant implications for optimizing Treg-derived exosome-based therapies for inflammation-related diseases.
Collapse
Affiliation(s)
- Simei Yue
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lingjiao Gong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yulin Tan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiaodan Zhang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Fei Liao
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Wuhan University Shenzhen Research Institute, Shenzhen, Guangdong 518000, China; Central Laboratory of Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
4
|
Gaspar LS, Pyakurel S, Xu N, D'Souza SP, Koritala BSC. Circadian Biology in Obstructive Sleep Apnea-Associated Cardiovascular Disease. J Mol Cell Cardiol 2025; 202:116-132. [PMID: 40107345 DOI: 10.1016/j.yjmcc.2025.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/16/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
A dysregulated circadian system is independently associated with both Obstructive Sleep Apnea (OSA) and cardiovascular disease (CVD). OSA and CVD coexistence is often seen in patients with prolonged untreated OSA. However, the role of circadian dysregulation in their relationship is unclear. Half of the human genes, associated biological pathways, and physiological functions exhibit circadian rhythms, including blood pressure and heart rate regulation. Mechanisms related to circadian dysregulation and heart function are potentially involved in the coexistence of OSA and CVD. In this article, we provide a comprehensive overview of circadian dysregulation in OSA and associated CVD. We also discuss feasible animal models and new avenues for future research to understand their relationship. Oxygen-sensing pathways, inflammation, dysregulation of cardiovascular processes, oxidative stress, metabolic regulation, hormone signaling, and epigenetics are potential clock-regulated mechanisms connecting OSA and CVD.
Collapse
Affiliation(s)
- Laetitia S Gaspar
- Centre for Neuroscience and Cell Biology, University of Coimbra, Portugal; Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal
| | - Santoshi Pyakurel
- Division of Pediatric Otolaryngology-Head and Neck Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| | - Na Xu
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | - Shane P D'Souza
- Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| | - Bala S C Koritala
- Division of Pediatric Otolaryngology-Head and Neck Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America; Department of Otolaryngology-Head and Neck Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America.
| |
Collapse
|
5
|
Ji LL. Nuclear factor κB signaling revisited: Its role in skeletal muscle and exercise. Free Radic Biol Med 2025; 232:158-170. [PMID: 40010515 DOI: 10.1016/j.freeradbiomed.2025.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 01/27/2025] [Accepted: 02/10/2025] [Indexed: 02/28/2025]
Abstract
Nuclear factor (NF) κB as a redox sensitive, anti-apoptotic and pro-inflammatory signaling molecule has been studied extensively for more than three decades. Its role in inducing antioxidant enzymes, defending against extracellular and intracellular stress and maintaining redox homeostasis in skeletal muscle has also been recognized. New research continues to explore the polytropic nature of NFκB in cellular function, especially its crosstalk with other important signaling pathways. Understanding of the broad impact of these functions has significant implications in health and disease of skeletal muscle as an organ designed for contraction and mobility. Two important aspects of muscle wellbeing, i.e., disease and aging, are not discussed in this review. This review will provide an update on the new findings related to NFκB involvement in multiple signaling pathways and refresh our knowledge of its activation in skeletal muscle with a special reference to physical exercise.
Collapse
Affiliation(s)
- Li Li Ji
- The Laboratory of Physiological Hygiene and Exercise Science, School of Kinesiology, University of Minnesota Twin Cities, USA.
| |
Collapse
|
6
|
Xu Y, Liu Z, Xu J, Xu L, He Z, Liu F, Wang Y. Role of brain-derived neurotrophic factor in frailty: From mechanisms to interventions. Biomed Pharmacother 2025; 186:118016. [PMID: 40187046 DOI: 10.1016/j.biopha.2025.118016] [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: 12/08/2024] [Revised: 03/23/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025] Open
Abstract
Frailty is a common medical syndrome which largely increases the risk of disability, depression, falls, hospitalization and mortality. An increasing number of research suggests that frailty is reversible by medical interventions at its early stage. Therefore, efficient detection is utterly important for frail population. Since numerous biological processes have been indicated in frail population, the critical regulators in these biological processes could provide biomarkers for early detection or treatment for frailty. The brain-derived neurotrophic factor (BDNF) has been associated with several biological process ranging from cognitive function to inflammation, therefore it could be an important regulator for frailty. In this review, we would discuss the mechanism association between different indicators of frailty and BDNF. Furthermore, we summarize the approaches to interfere with BDNF in healthy and pathologic condition, which could lead to identification of potential interventional strategies for frailty.
Collapse
Affiliation(s)
- Yuanchun Xu
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Ziyan Liu
- Department of Nursing, Traditional Chinese Medicine Hospital of Tongliang, Tongliang Chongqing 402560, China
| | - Jiao Xu
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Lunshan Xu
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Zongsheng He
- Department of Gastroenterology, Daping Hospital,Army Medical University, Chongqing 400042, China
| | - Fang Liu
- Department of Nursing, Traditional Chinese Medicine Hospital of Tongliang, Tongliang Chongqing 402560, China.
| | - Yaling Wang
- Department of Nursing, Daping Hospital, Army Medical University, Chongqing 400042, China.
| |
Collapse
|
7
|
Li X, Zhao Z, Ye H, Li D, Huang X, Lee JH, Liu R. CDO1 phosphorylation is required for IL-6-induced tumor cell proliferation through governing cysteine availability. Cell Commun Signal 2025; 23:194. [PMID: 40269955 DOI: 10.1186/s12964-025-02189-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 04/04/2025] [Indexed: 04/25/2025] Open
Abstract
Inflammatory pathways are often hijacked by cancer cells to favor their own proliferation and survival. Cysteine dioxygenase type 1 (CDO1), an iron-dependent thiol dioxygenase enzyme, catalyzes the rate-limiting step for cysteine oxidation, and so that functions as an important regulator of cellular cysteine availability. However, whether inflammatory environment affects CDO1 activity and cysteine oxidation and its potential impact on tumor growth remains substantially elusive. In the present study, we demonstrate that CDO1 activity and cysteine oxidation is inhibited upon IL-6 treatment, without noticeable alterations in CDO1 expression. Mechanistically, AKT1 phosphorylates CDO1 T89 under IL-6 treatment, which represses CDO1 enzymatic activity by disrupting iron incorporation. Further, AKT1-mediated CDO1 T89 phosphorylation is required for IL-6-elicited oral squamous cell carcinoma (OSCC) growth, and is associated with the progression of OSCC development. The present data discover a new mechanism by which AKT1-mediated CDO1 T89 phosphorylation governs cysteine oxidation to support OSCC growth, thereby highlighting its value as a potential anti-tumor target.
Collapse
Affiliation(s)
- Xin Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhe Zhao
- The Second Affiliated Hospital of Chengdu Medical College Nuclear Industry 416 Hospital, Chengdu, China
| | - Hongping Ye
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Dan Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiaoke Huang
- Department of Urology, Xindu District People's Hospital of Chengdu, Chengdu, 610500, People's Republic of China.
| | - Jong-Ho Lee
- Department of Health Sciences, The Graduate School of Dong-a University, Busan, 49315, Republic of Korea.
| | - Rui Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
8
|
Ren J, Yan G, Yang L, Kong L, Guan Y, Sun H, Liu C, Liu L, Han Y, Wang X. Cancer chemoprevention: signaling pathways and strategic approaches. Signal Transduct Target Ther 2025; 10:113. [PMID: 40246868 PMCID: PMC12006474 DOI: 10.1038/s41392-025-02167-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/01/2024] [Accepted: 02/04/2025] [Indexed: 04/19/2025] Open
Abstract
Although cancer chemopreventive agents have been confirmed to effectively protect high-risk populations from cancer invasion or recurrence, only over ten drugs have been approved by the U.S. Food and Drug Administration. Therefore, screening potent cancer chemopreventive agents is crucial to reduce the constantly increasing incidence and mortality rate of cancer. Considering the lengthy prevention process, an ideal chemopreventive agent should be nontoxic, inexpensive, and oral. Natural compounds have become a natural treasure reservoir for cancer chemoprevention because of their superior ease of availability, cost-effectiveness, and safety. The benefits of natural compounds as chemopreventive agents in cancer prevention have been confirmed in various studies. In light of this, the present review is intended to fully delineate the entire scope of cancer chemoprevention, and primarily focuses on various aspects of cancer chemoprevention based on natural compounds, specifically focusing on the mechanism of action of natural compounds in cancer prevention, and discussing in detail how they exert cancer prevention effects by affecting classical signaling pathways, immune checkpoints, and gut microbiome. We also introduce novel cancer chemoprevention strategies and summarize the role of natural compounds in improving chemotherapy regimens. Furthermore, we describe strategies for discovering anticancer compounds with low abundance and high activity, revealing the broad prospects of natural compounds in drug discovery for cancer chemoprevention. Moreover, we associate cancer chemoprevention with precision medicine, and discuss the challenges encountered in cancer chemoprevention. Finally, we emphasize the transformative potential of natural compounds in advancing the field of cancer chemoprevention and their ability to introduce more effective and less toxic preventive options for oncology.
Collapse
Affiliation(s)
- Junling Ren
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Guangli Yan
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Le Yang
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China
| | - Ling Kong
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Yu Guan
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Hui Sun
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China.
| | - Chang Liu
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Lei Liu
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Ying Han
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Xijun Wang
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China.
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China.
| |
Collapse
|
9
|
Škorjanc A, Smrkolj V, Umek N. GOReverseLookup: A gene ontology reverse lookup tool. Comput Biol Med 2025; 191:110185. [PMID: 40239235 DOI: 10.1016/j.compbiomed.2025.110185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 03/27/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND AND OBJECTIVE The Gene Ontology (GO) project has been pivotal in providing a structured framework for characterizing genes and annotating them to specific biological concepts. While traditional gene annotation primarily focuses on mapping genes to GO terms, descriptors of biological concepts, there is a growing need for tools facilitating reverse querying. This paper introduces GOReverseLookup, a novel tool designed to identify over- or underrepresented genes in researcher-defined states of interest (phenotypes), described by sets of GO terms. GOReverseLookup supplements the existing power of Gene Ontology by the possibility of orthologous gene querying across several databases, such as Ensembl and UniProtKB. This combination allows for a more nuanced identification of significant genes across a range of cross-species research contexts. METHODS GOReverseLookup queries genes associated with input GO terms. Bundles of GO terms encapsulate user-defined states of interest, e.g., angiogenesis. In the second stage of the analysis, all GO terms associated with each gene are fetched, and finally, the statistical relevance of the genes being involved in one (or all) of the defined states of interests is computed. RESULTS The two presented use cases illustrate its utility in discovering genes related to rheumatoid arthritis and genes linked with chronic inflammation and tumorigenesis. In both cases, GOReverseLookup discovered a substantial number of genes significantly associated with the aforementioned states of interest. CONCLUSIONS GOReverseLookup proves to be a valuable resource for unraveling the genetic basis of phenotypes, with diverse practical potentials in functional genomics, systems biology, and drug discovery. We anticipate that GOReverseLookup will significantly aid in identifying potential gene targets during the initial research phases.
Collapse
Affiliation(s)
- Aljoša Škorjanc
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Korytkova 2, Ljubljana, Slovenia
| | - Vladimir Smrkolj
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Korytkova 2, Ljubljana, Slovenia; National Institute of Chemistry, Hajdrihova ulica 19, Ljubljana, Slovenia
| | - Nejc Umek
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Korytkova 2, Ljubljana, Slovenia.
| |
Collapse
|
10
|
Sahoo R, Pattnaik S, Mohanty B, Mir SA, Behera B. Aryl hydrocarbon receptor (AHR) signalling: A double-edged sword guiding both cancer progression and cancer therapy. Biochim Biophys Acta Gen Subj 2025; 1869:130805. [PMID: 40222634 DOI: 10.1016/j.bbagen.2025.130805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/21/2025] [Accepted: 04/05/2025] [Indexed: 04/15/2025]
Abstract
Aryl Hydrocarbon Receptor (AHR) reported to be associated with major carcinogenic signalling cascades which cause cell proliferations, metastasis and invasion as well as immune imbalance. AHR Participates in cellular processes not only through genomic pathways to cause genomic alterations but also via nongenomic pathways to alter various cytoplasmic proteins. In addition, AHR senses a wide range of ligands that modulate its downstream mechanisms that are intricated in cancer induction and prevention. Thus, AHR functions as a two-sided sword where some AHR ligands contribute to enhance cancer whereas few are useful for cancer treatment. Therefore, AHR represent as a regulatory point in cancer progression and treatment. There is a need to reinvestigate the regulatory role of AHR in major intracellular pathways and to explore the potential of AHR ligand for the design of cancer therapeutics. This review emphasizes the interaction of AHR with pro-carcinogenic signalling pathways that modulate cancer induction and progression. Furthermore, it also discusses about the current discovery of AHR ligands for cancer initiation or inhibition. This information could be useful for development of therapeutic strategies for the management of cancer by targeting AHR.
Collapse
Affiliation(s)
- Rahul Sahoo
- Department of Biotechnology and Bioinformatics, Sambalpur University, Jyoti Vihar, Burla, Odisha 768019, India
| | - Sriya Pattnaik
- Department of Biotechnology and Bioinformatics, Sambalpur University, Jyoti Vihar, Burla, Odisha 768019, India
| | - Biswajit Mohanty
- Department of Biotechnology and Bioinformatics, Sambalpur University, Jyoti Vihar, Burla, Odisha 768019, India
| | - Showkat Ahmad Mir
- School of Life Sciences, Sambalpur University, Jyoti Vihar, Burla, Odisha 768019, India
| | - Birendra Behera
- Department of Biotechnology and Bioinformatics, Sambalpur University, Jyoti Vihar, Burla, Odisha 768019, India.
| |
Collapse
|
11
|
Cui J, Li H, Zhang T, Lin F, Chen M, Zhang G, Feng Z. Research progress on the mechanism of curcumin anti-oxidative stress based on signaling pathway. Front Pharmacol 2025; 16:1548073. [PMID: 40260389 PMCID: PMC12009910 DOI: 10.3389/fphar.2025.1548073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/24/2025] [Indexed: 04/23/2025] Open
Abstract
Oxidative stress refers to an imbalance between oxidative capacity and antioxidant capacity, leading to oxidative damage to proteins, lipids, and DNA, which can result in cell senescence or death. It is closely associated with the occurrence and development of various diseases, including cardiovascular diseases, nephropathy, malignant tumors, neurodegenerative diseases, hypertension, diabetes, and inflammatory diseases. Curcumin is a natural polyphenol compound of β-diketone, which has a wide range of pharmacological activities such as anti-inflammatory, antibacterial, anti-oxidative stress, anti-tumor, anti-fibrosis, and hypolipidemic, demonstrating broad research and development value. It has a wide range of biological targets and can bind to various endogenous biomolecules. Additionally, it maintains the redox balance primarily by scavenging ROS, enhancing the activity of antioxidant enzymes, inhibiting lipid peroxidation, and chelating metal ions. This paper systematically describes the antioxidative stress mechanisms of curcumin from the perspective of signaling pathways, focusing on the Keap1-Nrf2/ARE, NF-κB, NOX, MAPK and other pathways. The study also discusses potential pathway targets and the complex crosstalk among these pathways, aiming to provide insights for further research on curcumin's antioxidant mechanisms and its clinical applications.
Collapse
Affiliation(s)
- Jie Cui
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Haonan Li
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tianyi Zhang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Fengli Lin
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Meiyun Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Guimin Zhang
- Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Zhong Feng
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
- Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| |
Collapse
|
12
|
Liu W, Xu J, Chen L, Zhang D, Zhang J, Lu L, Zhang X, Huang X, Zhang G. Discovery of a novel BCL-2 inhibitor GW806742X for the treatment of TNBC. Biochem Pharmacol 2025; 237:116925. [PMID: 40199403 DOI: 10.1016/j.bcp.2025.116925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 03/24/2025] [Accepted: 04/02/2025] [Indexed: 04/10/2025]
Abstract
Triple-negative breast cancer(TNBC) lacks estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor (HER2), and traditional treatments cannot accurately target TNBC. Anthracycline- and paclitaxel-based chemotherapeutic agents are still the mainstay of treatment for TNBC, but these chemotherapeutic agents have major toxic side effects and are prone to drug resistance during the treatment of TNBC. In this study, we investigated the efficacy and potential mechanisms of action of GW806742X in the treatment of TNBC. We screened a library of 600 FDA-approved small molecule compounds to identify GW806742X, a small molecule that inhibits the viability of triple-negative breast cancer cells. In vitro, GW806742X was found to be cytotoxic to TNBC cells in a dose-dependent manner and to inhibit the growth of MDA-MB-468 tumors in vivo. Mechanistically, we used PharmMapper to predict the possible targets of GW806742X and demonstrated that the small molecule drug could directly bind to BCL-2 by molecular docking simulations and ITC experiments. Western blot analysis demonstrated that GW806742X could reduce BCL-2 protein levels and possibly promote BCL-2 degradation through the lysosomal pathway. Moreover, GW806742X disrupts NF-κB signaling. In conclusion, this study demonstrates that GW806742X can be a potential therapeutic agent for triple-negative breast cancer by targeting BCL-2.
Collapse
Affiliation(s)
- Wenjun Liu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China; School of Life Science, Soochow University, China
| | - Jia Xu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China; School of Life Science, Soochow University, China
| | - Li Chen
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Dongze Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Juan Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Linlin Lu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Xiaoming Zhang
- Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, China.
| | - Xue Huang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China.
| | - Guangbo Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China.
| |
Collapse
|
13
|
Liao X, Si H, Lai Y, Zhang X, Feng Y, Zhou T, Feng Y, Yu L. Porphyromonas gingivalis-OMVs promote the epithelial-mesenchymal transition of oral squamous cell carcinoma by inhibiting ferroptosis through the NF-κB pathway. J Oral Microbiol 2025; 17:2482924. [PMID: 40206095 PMCID: PMC11980236 DOI: 10.1080/20002297.2025.2482924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/10/2025] [Accepted: 03/17/2025] [Indexed: 04/11/2025] Open
Abstract
Background Recent studies reported the role of Porphyromonas gingivalis (P. g) in promoting oral squamous cell carcinoma (OSCC) progression. However, the molecular mechanism remains unclear. Materials and methods P. g-OMVs were isolated using ultracentrifugation method and characterized by transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA). CCK-8, migration, invasion, Quantitative real-time Polymerase Chain Reaction (qRT-PCR) and immunocytochemistry assays were performed to evaluate the effect of P. g-OMVs on tumor cells' proliferation, migration, invasion, epithelial-mesenchymal transition (EMT), and ferroptosis in vitro. Western blot was performed to study the phosphorylation of transcription factor nuclear factor kappa B (NF-κB). In vivo, the effect of P. g-OMVs on the growth of OSCC was evaluated using a xenograft tumor model, followed by hematoxylin and eosin and immunohistochemistry staining. Results TEM and NTA demonstrated that P. g-OMVs have a vesicular structure with a particle size of around 118 nm. Compared to the control group, P. g-OMVs significantly enhance the proliferation, migration, and invasion of tumor cells. In addition, P. g-OMVs promote the EMT of OSCC cells, which can be attenuated by ferroptosis activator erastin. Moreover, P. g-OMVs inhibit feroptosis of OSCC by activating NF-κB signaling. In vivo, P. g-OMVs significantly enhance tumor growth of OSCC. Inhibition of NF-κB could significnatly reduce the growth of OSCC, which can be further rescued using ferroptosis inhibitor Ferrostain-1. Conclusions P. g-OMVs promote OSCC progression by modulating the ferroptosis-related EMT through NF-κB signaling.
Collapse
Affiliation(s)
- Xinyue Liao
- Department of Pediatric Dentistry, The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, China
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
| | - Hang Si
- Department of Pediatric Dentistry, The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, China
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
| | - Yongxian Lai
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
- Department of Preventive Health Care, The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, China
| | - Xiaoyan Zhang
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
- Department of Periodontics & Oral Mucosal Diseases, The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, China
| | - Yun Feng
- Department of Pediatric Dentistry, The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, China
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
| | - Tiejun Zhou
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yan Feng
- Department of Pediatric Dentistry, The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, China
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
| | - Li Yu
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
- Department of Periodontics & Oral Mucosal Diseases, The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, China
- NHC Key Laboratory of Nuclear Technology Medical Transformation (Mianyang Central Hospital), Mianyang, China
| |
Collapse
|
14
|
Kharitonova EV, Sun Q, Ockerman F, Chen B, Zhou LY, Hysong MR, Tuftin B, Cao H, Mathias RA, Auer PL, Ober C, Raffield LM, Reiner AP, Cox NJ, Kelada SNP, Tao R, Li Y. EndoPRS: Incorporating endophenotype information to improve polygenic risk scores for clinical endpoints-A study in asthma. Am J Hum Genet 2025:S0002-9297(25)00107-7. [PMID: 40203832 DOI: 10.1016/j.ajhg.2025.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 04/11/2025] Open
Abstract
Polygenic risk score (PRS) prediction of complex diseases can be improved by leveraging related phenotypes. This has motivated the development of several multi-trait PRS methods that jointly model genetically correlated traits. However, these methods do not account for vertical pleiotropy, where one trait acts as a mediator for another. Here, we introduce endoPRS, a weighted lasso model that incorporates information from relevant endophenotypes to improve disease risk prediction without making assumptions about the genetic architecture underlying the endophenotype-disease relationship. Through extensive simulation analysis, we demonstrate the robustness of endoPRS in a variety of complex genetic frameworks. We also apply endoPRS to predict the risk of childhood-onset asthma in UK Biobank and All of Us by leveraging a paired genome-wide association study of eosinophil count, a relevant endophenotype. We find that endoPRS significantly improves prediction and transferability compared to many existing PRS methods, including multi-trait PRS methods MTAG and wMT-BLUP, which suggests advantages of endoPRS in real-life clinical settings.
Collapse
Affiliation(s)
- Elena V Kharitonova
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Quan Sun
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Center for Computation and Genomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Franklin Ockerman
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Brian Chen
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Laura Y Zhou
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Micah R Hysong
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bjoernar Tuftin
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hongyuan Cao
- Department of Statistics, Florida State University, Tallahassee, FL 32306, USA
| | - Rasika A Mathias
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Paul L Auer
- Division of Biostatistics, Data Science Institute, and Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alexander P Reiner
- Department of Epidemiology, University of Washington, Seattle, WA 98105, USA
| | - Nancy J Cox
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Samir N P Kelada
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ran Tao
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Yun Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
15
|
Wang G, Yang Q, Han Y, Zhang Y, Pan W, Ma Z, Tian H, Qu X. miR-32-5p suppresses the progression of hepatocellular carcinoma by regulating the GSK3β/NF-κB signaling. Acta Biochim Biophys Sin (Shanghai) 2025. [PMID: 40170617 DOI: 10.3724/abbs.2025038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly fatal form of malignancy that seriously threatens patient survival. The global 5-year survival rate for HCC patients ranges from 15% to 19%, and nearly 80% of patients are diagnosed at an advanced stage. Therefore, exploring the mechanism of HCC development and identifying biomarkers and therapeutic targets for HCC are vital. MicroRNAs (miRNAs), a class of noncoding single-stranded RNAs, are 20-24 nucleotides (nt) long. They play pivotal roles in modulating the progression of diverse diseases. The specific role of miR-32-5p in the development of HCC remains unclear. In this study, qRT-PCR is utilized to precisely determine the downregulated expression levels of miR-32-5p in HCC. Subsequently, functional analysis reveals the suppressive role of miR-32-5p in modulating the proliferative and migratory capabilities of HCC cells. Glycogen synthase kinase 3β (GSK3β) has emerged as a potential target of miR-32-5p, which is confirmed through a dual-luciferase reporter assay. Notably, the expression of GSK3β in HCC tissue specimens is negatively correlated with the abundance of miR-32-5p, and patients with high GSK3β expression have shorter survival time. Furthermore, the targeted downregulation of GSK3β remarkably impedes the proliferation and migration of tumor cells. This study suggests that miR-32-5p inhibits the proliferation and migration of HCC through regulating the GSK3β/NF-κB signaling pathway. Therefore, this study reveals that miR-32-5p exerts its suppressive effect on HCC progression, suggesting that it is a promising target for both diagnostic and targeted therapeutic interventions against HCC.
Collapse
Affiliation(s)
- Guangzhi Wang
- Department of Oncology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Qianqian Yang
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Yaqi Han
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Yunlong Zhang
- School of Medical Imaging, Shandong Second Medical University, Weifang 261053, China
| | - Wei Pan
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Zhongliang Ma
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Hui Tian
- Department of Geriatrics, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Xudong Qu
- Department of Intervention Radiology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| |
Collapse
|
16
|
Harada K, Sakamoto N, Kitaoka T, Nakamura Y, Kondo R, Morisue R, Hashimoto H, Yamamoto Y, Ukai S, Maruyama R, Sakashita S, Kojima M, Tanabe K, Ohdan H, Shitara K, Kinoshita T, Ishii G, Yasui W, Ochiai A, Ishikawa S. PI3 expression predicts recurrence after chemotherapy with DNA-damaging drugs in gastric cancer. J Pathol 2025; 265:472-485. [PMID: 39980125 PMCID: PMC11880974 DOI: 10.1002/path.6400] [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: 09/03/2024] [Revised: 12/10/2024] [Accepted: 01/06/2025] [Indexed: 02/22/2025]
Abstract
Despite recent advances in gastric cancer therapy, chemotherapy resistance and lack of methods for selecting combination regimens remain major problems. Organoids, which provide a culture system that more closely resembles tumor cell organization than traditional cell lines, can be established from surgical specimens with a high success rate and are widely used for drug sensitivity assays. In this study, we aimed to identify a novel biomarker for predicting multidrug resistance using gastric cancer organoids (GCOs). We evaluated 5-fluorouracil or oxaliplatin-resistant GCOs to find novel biomarkers that reflect multidrug resistance in gastric cancer. To examine the resistance mechanisms, RNA-sequencing analysis and ex vivo drug sensitivity testing were performed. The association of biomarkers with patient prognosis and chemotherapy efficacy was evaluated using three original cohorts with a total of 230 cases. The results were also validated with two independent public cohorts and single-cell RNA sequence data. Increased expression of peptidase inhibitor 3 (PI3) was detected in all 5-fluorouracil or oxaliplatin-resistant GCOs. Our findings suggest a potential association of PI3 expression with ribosome biosynthesis and RNA metabolism under organoid conditions. We also found that PI3 overexpression promoted 5-fluorouracil/oxaliplatin/cisplatin resistance but not paclitaxel resistance. Immunohistochemical evaluation of PI3 expression revealed that the PI3-positive gastric cancer group had a poorer outcome, especially in terms of time to recurrence. PI3 positivity was also an independent predictor of relapse after chemotherapy with DNA-damaging agents. PI3 promotes DNA-damaging drug resistance through multiple downstream regulations related to RNA and ribosomal metabolism. PI3 may be useful as a biomarker for the therapeutic selection of non-DNA-damaging agents. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Kenji Harada
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
- Department of Molecular Pathology, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Naoya Sakamoto
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
- Department of Molecular Pathology, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
- Department of Pathology and Clinical LaboratoryNational Cancer Center Hospital EastKashiwaJapan
| | - Takumi Kitaoka
- Department of Pathology and Clinical LaboratoryNational Cancer Center Hospital EastKashiwaJapan
- The Department of Pathology, Faculty of MedicineYamagata UniversityYamagataJapan
| | - Yuka Nakamura
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
| | - Ryotaro Kondo
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
| | - Ryo Morisue
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
- Department of Hepatobiliary and Pancreatic SurgeryNational Cancer Center Hospital EastKashiwaJapan
| | - Hiroko Hashimoto
- Division of Innovative Pathology and Laboratory MedicineExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
| | - Yusuke Yamamoto
- Division of Molecular and Cellular MedicineNational Cancer Center Research InstituteTokyoJapan
| | - Shoichi Ukai
- Department of Molecular Pathology, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Ryota Maruyama
- Department of Molecular Pathology, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Shingo Sakashita
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
- Department of Pathology and Clinical LaboratoryNational Cancer Center Hospital EastKashiwaJapan
| | - Motohiro Kojima
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
- Department of Pathology and Clinical LaboratoryNational Cancer Center Hospital EastKashiwaJapan
| | - Kazuaki Tanabe
- Department of Perioperative and Critical Care Management, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Kohei Shitara
- Department of Gastroenterology and Gastrointestinal OncologyNational Cancer Center Hospital EastKashiwaJapan
| | - Takahiro Kinoshita
- Division of Gastric SurgeryNational Cancer Center Hospital EastKashiwaJapan
| | - Genichiro Ishii
- Department of Pathology and Clinical LaboratoryNational Cancer Center Hospital EastKashiwaJapan
- Division of Innovative Pathology and Laboratory MedicineExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
| | - Wataru Yasui
- Department of Molecular Pathology, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Atsushi Ochiai
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
| | - Shumpei Ishikawa
- Division of PathologyExploratory Oncology Research & Clinical Trial Center, National Cancer CenterKashiwaJapan
- Department of Preventive Medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| |
Collapse
|
17
|
Tokumasu M, Sato A, Ito-Kureha T, Yamamoto M, Ohmine N, Semba K, Inoue JI, Yamamoto T. Tob negatively regulates NF-κB activation in breast cancer through its association with the TNF receptor complex. Cancer Gene Ther 2025:10.1038/s41417-025-00897-6. [PMID: 40169858 DOI: 10.1038/s41417-025-00897-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 02/23/2025] [Accepted: 03/21/2025] [Indexed: 04/03/2025]
Abstract
NF-κB mediates transcriptional regulation crucial to many biological functions, and elevated NF-κB activity leads to autoimmune and inflammatory diseases, as well as cancer. Since highly aggressive breast cancers have few therapeutic molecular targets, clarification of key molecular mechanisms of NF-κB signaling would facilitate the development of more effective therapy. In this report, we show that Tob, a member of the Tob/BTG family of antiproliferative proteins, acts as a negative regulator of the NF-κB signal in breast cancer. Studies with 35 human breast cancer cell lines reveal that Tob expression is negatively correlated with NF-κB activity. Analysis of The Cancer Genome Atlas (TCGA) database of clinical samples reveals an inverse correlation between Tob expression and NF-κB activity. Tob knockdown in human breast cancer cells promoted overactivation of NF-κB upon TNF-α treatment, whereas overexpression of Tob inhibited TNF-α stimulation-dependent NF-κB activation. Mechanistically, Tob associates with the TNF receptor complex I and consequently inhibits RIPK1 polyubiquitylation, leading to possible prevention of overwhelming activation of NF-κB.
Collapse
Affiliation(s)
- Miho Tokumasu
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan.
- Department of Immunology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan.
| | - Atsuko Sato
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Taku Ito-Kureha
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mizuki Yamamoto
- Research Center for Asian Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Nao Ohmine
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Kentaro Semba
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan
| | - Jun-Ichiro Inoue
- The University of Tokyo Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), Tokyo, Japan
| | - Tadashi Yamamoto
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan.
| |
Collapse
|
18
|
Ding Y, Yu Y. Therapeutic potential of flavonoids in gastrointestinal cancer: Focus on signaling pathways and improvement strategies (Review). Mol Med Rep 2025; 31:109. [PMID: 40017144 PMCID: PMC11884236 DOI: 10.3892/mmr.2025.13474] [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: 09/18/2024] [Accepted: 01/30/2025] [Indexed: 03/01/2025] Open
Abstract
Flavonoids are a group of polyphenolic compounds distributed in vegetables, fruits and other plants, which have considerable antioxidant, anti‑tumor and anti‑inflammatory activities. Several types of gastrointestinal (GI) cancer are the most common malignant tumors in the world. A large number of studies have shown that flavonoids have inhibitory effects on cancer, and they are recognized as a class of potential anti‑tumor drugs. Therefore, the present review investigated the molecular mechanisms of flavonoids in the treatment of different types of GI cancer and summarized the drug delivery systems commonly used to improve their bioavailability. First, the classification of flavonoids and the therapeutic effects of various flavonoids on human diseases were briefly introduced. Then, to clarify the mechanism of action of flavonoids on different types of GI cancer in the human body, the metabolic process of flavonoids in the human body and the associated signaling pathways causing five common types of GI cancer were discussed, as well as the corresponding therapeutic targets of flavonoids. Finally, in clinical settings, flavonoids have poor water solubility, low permeability and inferior stability, which lead to low absorption efficiency in vivo. Therefore, the three most widely used drug delivery systems were summarized. Suggestions for improving the bioavailability of flavonoids and the focus of the next stage of research were also put forward.
Collapse
Affiliation(s)
- Ye Ding
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yong Yu
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
19
|
Hu B, Yin Y, Zhang B, Li S, Li K, Zhou Y, Huang Q. Villin-1 regulates ferroptosis in colorectal cancer progression. FEBS J 2025; 292:1710-1725. [PMID: 39658274 DOI: 10.1111/febs.17350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/01/2024] [Accepted: 09/13/2024] [Indexed: 12/12/2024]
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer-related deaths worldwide. Despite extensive research, the mechanistic underpinnings driving CRC progression remain largely unknown. As a fundamental component of the brush border cytoskeleton, villin-1 (VIL1) acts as a marker for intestinal cell differentiation and maturation. Through a comprehensive transcriptomics analysis of eight studies (total sample: n = 1952), we consistently observed significant upregulation of VIL1 expression in CRC tumors compared with adjacent normal tissue. In our independent cohort, this notable upregulation has been further validated at both mRNA and protein levels in colon tumor tissues, relative not only to adjacent normal tissue but also to normal controls. Our data show that VIL1 promotes proliferation and migration while inhibiting apoptosis. Conversely, knockout of VIL1 suppresses proliferation and migration while inducing apoptosis. Mechanistically, we reveal that knocking out VIL1 activates ferroptosis and inhibits the migration of CRC cells, while overexpressing VIL1 yields the opposite effects, and vice versa. Additionally, VIL1 binds to Nuclear factor NF-kappa-B p105 subunit (NF-κB) and controls NF-κB expression. In vivo, overexpressing VIL1 inhibits ferroptosis, and induces the expression of NF-κB and lipocalin 2 (LCN2), thereby promoting CRC tumor growth. Thus, we have identified the VIL1/NF-κB axis as a pivotal regulator of CRC progression through ferroptosis modulation, unveiling VIL1 as a promising therapeutic target for CRC treatment via ferroptosis. Our study offers novel avenues for exploring the therapeutic potential of ferroptosis in CRC management, emphasizing the high potential of VIL1 in regulating colorectal tumorigenesis.
Collapse
Affiliation(s)
- Bangli Hu
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Yixin Yin
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Birong Zhang
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Siqi Li
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Kezhi Li
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - You Zhou
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Qinghua Huang
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
- Department of Breast Surgery, Wuzhou Red Cross Hospital, Wuzhou, China
| |
Collapse
|
20
|
Zheng H, Cao Z, Lv Y, Cai X. WTAP-mediated N6-methyladenine Modification of circEEF2 Promotes Lung Adenocarcinoma Tumorigenesis by Stabilizing CANT1 in an IGF2BP2-dependent Manner. Mol Biotechnol 2025; 67:1494-1508. [PMID: 38619801 DOI: 10.1007/s12033-024-01134-5] [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: 10/10/2023] [Accepted: 02/27/2024] [Indexed: 04/16/2024]
Abstract
N6-methyladenosine (m6A) is a common posttranscriptional RNA modification and plays an important role in cancer biology. Circular RNAs (circRNAs) are also reported to participate in lung adenocarcinoma (LUAD) progression. Here, we aimed to investigate the functions of Wilms tumor 1-associating protein (WTAP) methyltransferase and circEEF2 in LUAD cell tumorigenesis, and probe whether circEEF2 functioned through WTAP-induced m6A modification and its potential mechanisms. Functional analyses were conducted by tube formation, sphere formation, 5-ethynyl-2'-deoxyuridine (EdU), flow cytometry, and transwell assays in vitro as well as tumor formation experiments in mice, respectively. The N6-methyladenine (m6A) modification in circEEF2 mRNA was determined by RNA immunoprecipitation (Me-RIP) assay. The interaction between IGF2BP2 (Insulin Like Growth Factor 2 MRNA-Binding Protein 2) and circEEF2 or Calcium-activated nucleotidase 1 (CANT1) mRNA was confirmed using RIP assay. LUAD tissues and cells showed high circEEF2 expression, and the deficiency of circEEF2 suppressed LUAD cell angiogenesis, stemness, proliferation, migration, and invasion. WTAP induced circEEF2 m6A modification. WTAP silencing repressed the oncogenic phenotypes of LUAD cells via stabilizing circEEF2 in an m6A-dependent manner. IGF2BP2 interacted with circEEF2 and CANT1, and WTAP and circEEF2 could regulate CANT1 expression through IGF2BP2. The inhibition of LUAD cell oncogenic phenotypes caused by circEEF2 deficiency was abolished by CANT1 overexpression. In addition, WTAP silencing impeded LUAD growth via modulating circEEF2 and CANT1 in vivo. WTAP-mediated m6A modification of circEEF2 promotes lung adenocarcinoma growth and tumorigenesis by stabilizing CANT1 through IGF2BP2.
Collapse
Affiliation(s)
- Hao Zheng
- Department of Respiratory and Critical Care Medicine, Lishui People's Hospital, 15, Dazhong Street, Liandu District 323000, Lishui City, Zhejiang Province, China
| | - Zhuo Cao
- Department of Respiratory and Critical Care Medicine, Lishui People's Hospital, 15, Dazhong Street, Liandu District 323000, Lishui City, Zhejiang Province, China
| | - Yuankai Lv
- Department of Respiratory and Critical Care Medicine, Lishui People's Hospital, 15, Dazhong Street, Liandu District 323000, Lishui City, Zhejiang Province, China
| | - Xiaoping Cai
- Department of Respiratory and Critical Care Medicine, Lishui People's Hospital, 15, Dazhong Street, Liandu District 323000, Lishui City, Zhejiang Province, China.
| |
Collapse
|
21
|
Tajeri S, Langsley G. Virulence attenuation of Theileria annulata-transformed macrophages. Trends Parasitol 2025; 41:301-316. [PMID: 40057452 DOI: 10.1016/j.pt.2025.02.007] [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: 01/09/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 04/05/2025]
Abstract
Tropical theileriosis is a significant tick-borne disease affecting cattle. For decades an empirical live attenuated vaccine has been the primary method of controlling disease. The vaccine is produced through prolonged culture of Theileria annulata schizont-transformed macrophages, but how loss of virulence occurs remains unclear. Notably attenuated (vaccine) macrophages display dampened dissemination potential compared with their original, virulent counterparts. In addition, parasite schizonts in attenuated macrophages have significantly lost their ability to differentiate into merozoites. This review discusses the changes that occur during long-term passage of T. annulata-transformed bovine macrophages and how they contribute to loss of virulence, defined as heightened dissemination. Finally, we also suggest that a common parasite-dependent pathway is potentially involved in both macrophage dissemination and parasite merogony.
Collapse
Affiliation(s)
- Shahin Tajeri
- Laboratoire de Biologie des Apicomplexes, Faculté de Médecine, Université Paris Descartes - Sorbonne Paris Cité, Paris, France; INSERM U1016, CNRS UMR8104, Cochin Institute, Paris, France; Freie Universität Berlin, Institute for Parasitology and Tropical Veterinary Medicine, Berlin, Germany; Freie Universität Berlin, Veterinary Centre for Resistance Research, Berlin, Germany.
| | - Gordon Langsley
- Laboratoire de Biologie des Apicomplexes, Faculté de Médecine, Université Paris Descartes - Sorbonne Paris Cité, Paris, France; INSERM U1016, CNRS UMR8104, Cochin Institute, Paris, France.
| |
Collapse
|
22
|
Blanco LP, Salmeri N, Temkin SM, Shanmugam VK, Stratton P. Endometriosis and autoimmunity. Autoimmun Rev 2025; 24:103752. [PMID: 39828017 DOI: 10.1016/j.autrev.2025.103752] [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: 09/06/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Endometriosis is a female-specific chronic condition that affects 1 in 10 women and other individuals with a uterus worldwide with common symptoms that include pelvic pain and infertility. Reliable and effective non-invasive biomarkers for endometriosis do not exist, and therefore currently a diagnosis of endometriosis requires direct visualization of lesions at surgery. Similarly, few safe and effective management strategies exist for endometriosis, with hormonal interventions and surgery only providing temporary symptom control. The development of endometriosis involves the implantation and proliferation of ectopic endometrial cells which triggers local and systemic inflammation and fibrosis. While multiple genetic, environmental, and lifestyle factors appear to influence the natural history of endometriosis, chronic inflammation is a hallmark feature associated with development and progression of the disease. Data further shows that endometriosis commonly co-occurs with autoimmune diseases, adding evidence that immune dysfunction likely contributes to the pathogenesis of this disorder. Specific innate and adaptive immune system drivers of endometriosis remain to be identified and additional research is needed to elucidate the mechanistic underpinnings of this debilitating disease. In this narrative review, we discuss the shared biological mechanisms and plausible immune-related connections between endometriosis and autoimmunity.
Collapse
Affiliation(s)
- Luz P Blanco
- National Institute of Arthritis, and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Noemi Salmeri
- Office of Research on Women's Health, Office of the Director, National Institutes of Health, Bethesda, MD, United States of America
| | - Sarah M Temkin
- Office of Research on Women's Health, Office of the Director, National Institutes of Health, Bethesda, MD, United States of America
| | - Victoria K Shanmugam
- Office of Autoimmune Disease Research, Office of Research on Women's Health, Office of the Director, National Institutes of Health, Bethesda, MD, United States of America
| | - Pamela Stratton
- Office of Research on Women's Health, Office of the Director, National Institutes of Health, Bethesda, MD, United States of America; Scientific Consulting Group, Gaithersburg, MD, United States of America.
| |
Collapse
|
23
|
Sarsarshahi S, Bhattacharya S, Zacharias ZR, Kamel ES, Houtman JCD, Nejadnik R. Highly variable aggregation and glycosylation profiles and their roles in immunogenicity to protein-based therapeutics. J Pharm Sci 2025; 114:103771. [PMID: 40139530 DOI: 10.1016/j.xphs.2025.103771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
Production of antibodies against protein-based therapeutics (e.g., monoclonal antibodies (mAbs)) by a recipient's immune system can vary from benign symptoms to chronic neutralization of the compound, and in rare cases, a lethal cytokine storm. One critical factor that can induce or contribute to an anti-drug antibody (ADA) response is believed to be the presence of aggregated proteins in protein-based therapeutics. There is a high level of variability in the aggregation of different proteins, which adds to the complexity in understanding the immune response to these drugs. Furthermore, the level of glycosylation of proteins, which increases drug stability, functionality, and serum half-life, is highly variable and may influence their immunogenicity. Considering the abundance of literature on the effect of aggregation and glycosylation on the immunogenicity of protein-based therapeutics, this review aims to summarize the current knowledge and clarify the immunogenic effects of different protein-based therapeutics such as mAbs. This review focuses on the properties of aggregated proteins and elucidates their relationship with immunogenicity. The contribution of different immune cell subsets and the mechanisms in aggregation-induced immunogenicity are also reviewed. Finally, the potential effects of each glycan, such as sialic acid, mannose, and fucose, on protein-based therapeutics' immunogenicity and stability is discussed.
Collapse
Affiliation(s)
- Sina Sarsarshahi
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, United States
| | - Sanghati Bhattacharya
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, United States
| | - Zeb R Zacharias
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States; Human Immunology Core, University of Iowa, Iowa City, IA, United States
| | - Eman S Kamel
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, United States
| | - Jon C D Houtman
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States; Human Immunology Core, University of Iowa, Iowa City, IA, United States; Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
| | - Reza Nejadnik
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, United States.
| |
Collapse
|
24
|
Khan IM, Gul H, Khan S, Nassar N, Khalid A, Swelum AA, Wang Z. Green tea polyphenol epigallocatechin-3-gallate mediates an antioxidant response via Nrf2 pathway in heat-stressed poultry: A review. Poult Sci 2025; 104:105071. [PMID: 40157268 PMCID: PMC11995091 DOI: 10.1016/j.psj.2025.105071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/15/2025] [Accepted: 03/18/2025] [Indexed: 04/01/2025] Open
Abstract
Heat stress is a critical challenge in the poultry industry. It arises when birds are exposed to elevated ambient temperatures beyond their thermoneutral zone, often exacerbated by high humidity and inadequate ventilation. This condition disrupts the birds' ability to maintain thermal homeostasis, leading to physiological and behavioral changes such as increased panting, reduced feed intake, and elevated water consumption. These responses aim to dissipate heat but often result in energy imbalances, oxidative stress, and impaired immune function. Green tea polyphenols (GTPs) mitigate heat stress in poultry birds by modulating oxidative stress pathways, primarily by scavenging reactive oxygen species (ROS) and enhancing antioxidant defense mechanisms. These pathways play a pivotal role in neutralizing ROS generated during oxidative stress, inflammation, and exposure to electrophilic compounds. This action helps restore cellular balance and enhances overall antioxidant defense mechanisms by converting harmful free radicals into less reactive molecules, such as water and oxygen. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) plays a significant character in the activation of the enzymatic antioxidants network. It translocates to the nucleus upon activation, binds to antioxidant response elements (AREs) in the promoter regions of target genes, and upregulates the expression of key antioxidant enzymes. Therefore, the regulation of Nrf2 is considered a critical molecular marker in mitigating the effects of heat stress, as its activation enhances the expression of antioxidant and detoxification enzymes, protecting against oxidative damage and inflammation induced by elevated temperatures. This exploratory review summarizes the antioxidant mechanisms and anti-oxidative stress effects of GTPs in mitigating heat stress in poultry. It highlights the cytoprotective molecular basis of epigallocatechin-3-gallate (EGCG), particularly its role in modulating Nrf2-mediated cellular pathways, which enhance antioxidant defense systems and protect against oxidative damage.
Collapse
Affiliation(s)
| | - Haji Gul
- District Livestock Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Samiullah Khan
- The Scientific Observing and Experimental Station of Crop Pest in Guiyang, Ministry of Agriculture, Institute of Entomology, Guizhou University, Guiyang, China
| | - Nourhan Nassar
- College of Life Science, Anhui Agricultural University, Hefei, Anhui, China; Department of Clinical Pathology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh 13736, QG, Egypt
| | - Anam Khalid
- College of Life Science, Anhui Agricultural University, Hefei, Anhui, China
| | - Ayman A Swelum
- Department of Theriogenology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt.
| | - Zaigui Wang
- College of Life Science, Anhui Agricultural University, Hefei, Anhui, China.
| |
Collapse
|
25
|
Shen Z, Wang C, Cao C, Wang G, Li Z. Gastroesophageal reflux disease as a risk factor for oral cavity and pharyngeal cancer: a Mendelian randomization study. Discov Oncol 2025; 16:353. [PMID: 40100509 PMCID: PMC11920546 DOI: 10.1007/s12672-025-02105-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/10/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Previous observational studies have not clearly examined the impact of gastroesophageal reflux disease (GERD) on the risk of oral cavity and pharyngeal cancer (OCPC). To provide more evidence to elucidate this issue, we used Mendelian randomization (MR) to analyze the causal effect of GERD on OCPC and its subtypes. METHODS We obtained the summary data of genome-wide association studies (GWAS) of European ancestry to perform MR analysis. GERD was considered the exposure, and OCPC (subtypes include oral cavity cancer (OCC) and oropharynx cancer (OPC)) was defined as the outcome. We aimed to investigate whether GERD has a causal effect on OCPC. We then attempted to obtain more accurate causal estimates by correcting for potential confounders such as smoking behavior, drinking behavior, body mass index (BMI), and type 2 diabetes (T2D). We also performed extensive sensitivity analyses to assess the robustness of the primary analysis results. RESULTS Univariate MR analysis showed that GERD had a positive causal effect on OCPC (IVW: discovery, OR = 2.09 (95% CI 1.30-3.37), P = 0.0023; validation, OR = 1.90 (95% CI 1.26-2.87), P = 0.0020) and OCC (IVW: discovery, OR = 2.01 (95% CI: 1.21-3.33), P = 0.0066; validation, OR = 2.60 (95% CI 1.47-4.59), P = 0.0010). Although GERD increased the risk of OPC, this effect was statistically significant only in the discovery analysis (IVW: discovery, OR = 2.30 (95% CI 1.08-4.89), P = 0.0307; validation, OR = 1.15 (95% CI 0.67-1.97), P = 0.6199), the causal direction remained consistent. After adjusting for smoking, alcohol consumption, BMI, and T2D in multivariate analysis, the results remained largely consistent. CONCLUSIONS Our study showed that GERD significantly increased the overall risk of OCPC, and similar results were found for its subtype OCC. This causal effect appears to be independent of cigarette use, drinking habits, BMI, and T2D. However, evidence for a causal effect of GERD on OPC is limited, and further research is expected to extend this finding. Future studies should explore the specific biological mechanisms through which GERD increases OCPC risk.
Collapse
Affiliation(s)
- Zixiong Shen
- Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, 130000, China
| | - Chuanlei Wang
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, 130000, China
- Key Laboratory of General Surgery Health Department of Jilin Province, Changchun, 130000, China
| | - Chunli Cao
- Department of Stomatology, The First Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Guangyi Wang
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, 130000, China
- Key Laboratory of General Surgery Health Department of Jilin Province, Changchun, 130000, China
| | - Zhiqin Li
- Surgical Intensive Care Unit, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
26
|
Dai Z, Yang H, Yin P, Liu X, Zhang L, Dou Y, Sun S. Applications of Cyclodextrin-Based Drug Delivery Systems in Inflammation-Related Diseases. Pharmaceutics 2025; 17:378. [PMID: 40143041 PMCID: PMC11945956 DOI: 10.3390/pharmaceutics17030378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/24/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Currently, inflammation diseases are one of the leading causes of mortality worldwide. The therapeutic drugs for inflammation are mainly steroidal and non-steroidal anti-inflammatory drugs. However, the use of these anti-inflammatory drugs over a prolonged period is prone to causing serious side effects. Accordingly, it is particularly critical to design an intelligent target-specific drug delivery system to control the release of drugs in order to mitigate the side effects of anti-inflammatory drugs without limiting their activity. Meanwhile, cyclodextrin-based nano-delivery systems have garnered significant attention in contemporary pharmaceutical research owing to their capacity to enhance drug bioavailability, enable site-specific targeted accumulation, prolong the systemic circulation duration, facilitate synergistic therapeutic outcomes, and exhibit superior biocompatibility profiles. It is worth noting that cyclodextrin-based drug delivery systems show great potential in inflammation-related diseases. However, few studies have systematically reviewed their design strategies and application advancements. Here, we summarize the structural and chemical modification strategies of cyclodextrins, as well as cyclodextrin-based drug delivery systems and their applications in inflammation-related diseases. In summary, the aim is to provide a bit of insight into the development of cyclodextrin-based drug delivery systems for inflammation-related diseases.
Collapse
Affiliation(s)
- Zelan Dai
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming 650032, China;
- Department VII of Biological Products, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650031, China; (H.Y.); (P.Y.); (X.L.); (L.Z.); (Y.D.)
| | - Huijuan Yang
- Department VII of Biological Products, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650031, China; (H.Y.); (P.Y.); (X.L.); (L.Z.); (Y.D.)
| | - Peng Yin
- Department VII of Biological Products, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650031, China; (H.Y.); (P.Y.); (X.L.); (L.Z.); (Y.D.)
| | - Xingkang Liu
- Department VII of Biological Products, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650031, China; (H.Y.); (P.Y.); (X.L.); (L.Z.); (Y.D.)
| | - Ling Zhang
- Department VII of Biological Products, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650031, China; (H.Y.); (P.Y.); (X.L.); (L.Z.); (Y.D.)
| | - Youwei Dou
- Department VII of Biological Products, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650031, China; (H.Y.); (P.Y.); (X.L.); (L.Z.); (Y.D.)
| | - Shibo Sun
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming 650032, China;
| |
Collapse
|
27
|
Dutta D, Hoque AA, Paul B, Begum S, Sarkar UA, Mukherjee B. Molecular insights into the antineoplastic potential of apigenin and its derivatives: paving the way for nanotherapeutic innovations. Expert Opin Drug Deliv 2025:1-20. [PMID: 40063738 DOI: 10.1080/17425247.2025.2477664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 03/06/2025] [Indexed: 03/18/2025]
Abstract
INTRODUCTION Apigenin, a widely distributed bioactive flavonoid, has recently gained excellent attention among researchers as an effective anticancer drug that can alternate cancer-signaling pathways, induce programmed cell death, and reduce tumor growth in various cancer types. Despite its impressive anti-neoplastic activity, high hydrophobicity, and nonspecific biodistribution make apigenin difficult for pharmaceutical applications. AREAS COVERED We highlighted the therapeutic potential of apigenin and its derivatives in different cancer types, along with their mechanism of action. Nanoengineered drug delivery systems have remarkable applications in minimizing drug degradation and enhancing the therapeutic efficacy of drugs with sustained release, prolonged blood retention time, and reduced off-target toxicities. This review has evaluated and explored the molecular interactions of this novel flavonoid in various cancer signaling pathways to selectively inhibit neoplastic development in multiple cancer types. To ensure the complete coverage of the explored research area, Google Scholar, PubMed, and Web of Science were used to find not only the most relevant but also connected and similar articles. EXPERT OPINION A comprehensive overview of apigenin nanotherapy in cancer treatment can establish a platform to overcome its difficulties for pharmaceutical applications and efficient clinical translation from bench to bedside.
Collapse
Affiliation(s)
- Debasmita Dutta
- Dana Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Ashique Al Hoque
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Brahamacharry Paul
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Shahnaz Begum
- Department of Chemistry, Jadavpur University, Kolkata, India
| | - Uday Aditya Sarkar
- Dana Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Biswajit Mukherjee
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| |
Collapse
|
28
|
Zhang YX, Wan H, Shan GY, Cheng JY, Liu YY, Shi WN, Li HJ. Pharmacological role of Herba Patriniae and Coix seed in colorectal cancer. World J Gastrointest Oncol 2025; 17:99673. [PMID: 40092956 PMCID: PMC11866235 DOI: 10.4251/wjgo.v17.i3.99673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 10/31/2024] [Accepted: 12/25/2024] [Indexed: 02/14/2025] Open
Abstract
Colorectal cancer (CRC) is the most prevalent cancer globally, and its traditional treatment modalities commonly encompass radiation therapy, chemotherapy, surgery and the administration of cytotoxic drugs. Currently, novel chemotherapy drugs that combine traditional Chinese medicine (TCM) with herbal extracts exhibit superior comprehensive benefits. Herein, we delved into an article authored by Wang et al, focusing specifically on the pharmacological effects of "Herba Patriniae and Coix seed (HC)" and their targeted mechanisms in combating CRC. From the perspective of TCM philosophy, damp-heat stagnation and toxicity are the cardinal pathogenic factors underlying CRC. HC, renowned for their abilities to antipyretic and enhance diuresis, have demonstrated promising efficacy in preliminary studies for the treatment of CRC. These findings offer potential insights in favor of fostering anti-cancer medications.
Collapse
Affiliation(s)
- Yu-Xin Zhang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Hui Wan
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Guan-Yue Shan
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Jun-Ya Cheng
- Department of Bioengineering, Pharmacy School of Jilin University, Changchun 130061, Jilin Province, China
| | - Yi-Ying Liu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Wen-Na Shi
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Hai-Jun Li
- Institute of Liver Diseases, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| |
Collapse
|
29
|
Gao Y, Sun L, Qiao C, Liu Y, Wang Y, Feng R, Zhang H, Zhang Y. Cyclodextrin-based delivery systems for chemical and genetic drugs: Current status and future. Carbohydr Polym 2025; 352:123174. [PMID: 39843078 DOI: 10.1016/j.carbpol.2024.123174] [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: 12/01/2024] [Accepted: 12/18/2024] [Indexed: 01/24/2025]
Abstract
Cyclodextrins (CDs) are cyclic polysaccharides characterized by their unique hollow structure, making them highly effective carriers for pharmaceutical agents. CD-based delivery systems are extensively utilized to enhance drug stability, increase solubility, improve oral bioavailability, and facilitate controlled release and targeted delivery. This review initially provides a concise overview of nano drug delivery systems, followed by a detailed introduction of the structural features and benefits of CDs. It further summarizes the applications of CD-based delivery systems and offers insights for the rational design of drug delivery systems. In this review, CD-based delivery systems are categorized into several types, such as covalently modified CD derivatives, non-modified CD inclusion complexes, poly-cyclodextrins and others. The application of CD-based systems for the delivery of genetic therapeutic agents and co-delivery of gene and drug is also presented. Finally, this review discusses potential challenges and opportunities that may arise in the future. With the development of nanotechnology and optimization of preparation process, CD-based drug delivery systems will provide a more effective, precise and safe approach to drug therapy.
Collapse
Affiliation(s)
- Yikun Gao
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Le Sun
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Chu Qiao
- Department of Pharmacy, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Yuqing Liu
- Department of Pharmacy, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Yang Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Rui Feng
- School of Pharmacy, China Medical University, Shenyang 110122, China.
| | - Hong Zhang
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang 110016, China; Department of Pharmacy, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Youxi Zhang
- Department of Pharmacy, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| |
Collapse
|
30
|
Doğan S, Tuncer MC, Özdemir İ. Inhibition of Retinoblastoma Cell Growth by Boswellic Acid Through Activation of the Suppressing Nuclear Factor-κB Activation. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:480. [PMID: 40142291 PMCID: PMC11944177 DOI: 10.3390/medicina61030480] [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: 01/23/2025] [Revised: 03/02/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025]
Abstract
Background and Objectives: Despite the development of treatment methods and the emergence of alternative new approaches in recent years, the visual prognosis of retinoblastoma contains deficiencies and this situation increases the need for the development of new treatment approaches. The cytotoxic and apoptosis-inducing effects of the combination of boswellic acid (BA), which has been determined to have significant potential in preclinical and clinical studies of various diseases, and Cisplatin (Cis), a potent chemotherapy agent, were investigated on the human retinoblastoma cell line (Y79). Materials and Methods: The cytotoxic effect of BA and Cis on Y79 cells was determined by the water soluble tetrazolium-1 (WST-1) test, the apoptotic rate of the cells was determined by annexin V staining, and the gene expressions of Protein53 (p53), Caspase-3 and Nuclear factor kappa B (NF-κB), which play an important role in apoptosis, were determined by RT-qPCR analysis. Interleukin 1-beta (IL1-β), tumor necrosis factor-α (TNF-α) and interferon γ (IFN-γ) levels were analyzed in cell lysates obtained from the experimental groups. Results: The combination of BA and Cis selectively inhibited the growth of Y79 cells and modulated NF-κB signaling, potentially through post-translational regulatory mechanisms. Moreover, it induced apoptosis by increasing p53 and Caspase-3 expressions, confirming its pro-apoptotic effects. Additionally, the combination treatment was associated with a reduction in inflammatory cytokine levels (TNF-α, IL1-β), suggesting a potential regulatory effect on inflammation-related pathways rather than direct inhibition of NF-κB activation. Conclusions: These findings suggest that BA combined with Cis inhibits Y79 retinoblastoma cell growth by inducing apoptosis and modulating NF-κB signaling. While NF-κB mRNA levels increased, reduced inflammatory cytokines and enhanced apoptosis suggest potential post-translational regulation. Further studies are needed to confirm NF-κB protein-level effects and in vivo efficacy.
Collapse
Affiliation(s)
- Semih Doğan
- Department of Ophthalmology, Faculty of Medicine, Beykent University, İstanbul 34398, Turkey;
| | - Mehmet Cudi Tuncer
- Department of Anatomy, Faculty of Medicine, Dicle University, Diyarbakır 21280, Turkey
| | - İlhan Özdemir
- Department of Gynecology and Obstetrics, Faculty of Medicine, Atatürk University, Erzurum 25240, Turkey;
| |
Collapse
|
31
|
Zeyneb H, Song Y, Wang L, Zheng J, Wang W, Pei H, Cao X. Preventive effect of quinoa polysaccharides on lipopolysaccharide-induced inflammation in mice through gut microbiota regulation. Int J Biol Macromol 2025; 307:141899. [PMID: 40068754 DOI: 10.1016/j.ijbiomac.2025.141899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/28/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025]
Abstract
Inflammation significantly influences the development of gastrointestinal (GI) diseases such as inflammatory bowel diseases (IBD)and ulcerative colitis, which disrupts normal digestive functions, leading to tissue damage and various symptoms. This research explores the preventive effects of quinoa polysaccharides (QPS) on lipopolysaccharide (LPS)-induced systemic acute inflammation in mice and their mechanism of action. The findings revealed that QPS alleviated LPS-induced inflammation symptoms, enhanced the mice behavior score and their immune organ index, reduced pro-inflammatory cytokines (IL-6, TNF-α and IL-1β) levels, elevated the expression level of tight junction proteins (ZO-1, MUC2). Additionally, the levels of superoxide dismutase (SOD), malondialdehyde (MDA) and total antioxidant capacity (T-AOC) were improved via QPS administration. Further, our research suggested that QPS enhanced the diversity and abundance of gut microbiota compared to that of LPS mice, leading to an increase in the short-chain fatty acids in mice feces. Linear discriminant analysis (LDA) effect size (LEfSe) showed that QPS administration could lead to a range of gut biomarkers, promoting the enhancement of polysaccharide-metabolizing bacteria. The results of 16S rRNA sequencing indicated that QPS alleviates LPS-induced inflammation by enhancing the richness of beneficial bacteria such as Bacteroides and Lactobacillus. Linear discriminant analysis (LDA) effect size (LEfSe) showed that QPS administration could lead to a range of gut biomarkers, promoting the enhancement of polysaccharide-metabolizing bacteria. UPLC Q-TOF-MS was performed to analyze metabolites in the fecal samples. LPS administration significantly altered metabolite levels detected in mice feces in which some metabolites have decreased such as xanthosine and hypoxanthine while an increase in some metabolites in mice that received QPS, metabolomics analysis showed the beneficial effects of QPS primarily mediated via amino and bile acid-related metabolism pathways. Our research could offer the basis for further studies and applications of quinoa polysaccharides.
Collapse
Affiliation(s)
- Hitache Zeyneb
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology & Business University, Beijing 100048, China
| | - Ya Song
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology & Business University, Beijing 100048, China
| | - Lin Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology & Business University, Beijing 100048, China
| | - Jimin Zheng
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Wei Wang
- Institute of Analysis and Testing, Beijing Academy of Science and Technology (Beijing Center for Physical &Chemical Analysis), Beijing 100094, China
| | - Hairun Pei
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology & Business University, Beijing 100048, China.
| | - Xueli Cao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology & Business University, Beijing 100048, China.
| |
Collapse
|
32
|
Lang SQ, Kong JJ, Li GB, Liu J. Prognostic value of CRP-albumin-lymphocyte index in patients with intrahepatic cholangiocarcinoma after radical resection. Front Med (Lausanne) 2025; 12:1543665. [PMID: 40115790 PMCID: PMC11922830 DOI: 10.3389/fmed.2025.1543665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 02/24/2025] [Indexed: 03/23/2025] Open
Abstract
Purpose The aim of this study is to explore the prognostic value of CRP-Albumin-Lymphocyte (CALLY) index in patients undergoing radical resection of intrahepatic cholangiocarcinoma (ICC). Patients and methods Retrospectively collected clinical data of 286 patients with ICC who underwent radical surgery at Shandong Provincial Hospital from July 2010 to July 2021. Univariate and multivariate analyses were used to evaluate the correlation between the CALLY index and overall survival (OS) and recurrence-free survival (RFS), and a nomogram prediction model was established based on the results. The accuracy of the model was evaluated using concordance index (C-index), calibration curves, decision curve analysis (DCA), and the receiver operating characteristic (ROC) curve was used to compare the prognostic value of the nomogram model with the TNM staging system. Results The optimal cut-off value of CALLY index was 1.81. In the training set, multifactorial Cox regression analysis showed that CALLY index <1.81 was an independent risk factor for OS and RFS (p < 0.05). Compared to neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), systemic immune inflammation index (SII), and modified Glasgow prognostic score (mGPS), CALLY index had a higher area under the curve (AUC). The nomogram established based on the results of multifactorial analysis demonstrated strong efficacy in survival prediction, and the ROC curve showed that the nomogram had a higher prognostic value than TNM staging. Conclusion The CALLY index is independently associated with OS and RFS in patients after radical resection of ICC, and the nomogram model based on it shows significantly higher efficacy in predicting the long-term prognosis of patients after radical resection of ICC, and is more accurate than TNM staging.
Collapse
Affiliation(s)
- Shi-Qian Lang
- Department of Liver Transplantation and Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jun-Jie Kong
- Department of Liver Transplantation and Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Guang-Bing Li
- Department of Liver Transplantation and Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jun Liu
- Department of Liver Transplantation and Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
33
|
Li X, Zhang W, Fang Y, Sun T, Chen J, Tian R. Large-scale CRISPRi screens link metabolic stress to glioblastoma chemoresistance. J Transl Med 2025; 23:289. [PMID: 40050992 PMCID: PMC11887098 DOI: 10.1186/s12967-025-06261-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 02/14/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Glioblastoma (GBM) patients frequently develop resistance to temozolomide (TMZ), the standard chemotherapy. While targeting cancer metabolism shows promise, the relationship between metabolic perturbation and drug resistance remains poorly understood. METHODS We performed high-throughput CRISPR interference screens in GBM cells to identify genes modulating TMZ sensitivity. Findings were validated using multiple GBM cell lines, patient-derived glioma stem cells, and clinical data. Molecular mechanisms were investigated through transcriptome analysis, metabolic profiling, and functional assays. RESULTS We identified phosphoglycerate kinase 1 (PGK1) as a key determinant of TMZ sensitivity. Paradoxically, while PGK1 inhibition suppressed tumor growth, it enhanced TMZ resistance by inducing metabolic stress. This activated AMPK and HIF-1α pathways, leading to enhanced DNA damage repair through 53BP1. PGK1 expression levels correlated with TMZ sensitivity across multiple GBM models and patient samples. CONCLUSIONS Our study reveals an unexpected link between metabolic stress and chemoresistance, demonstrating how metabolic adaptation can promote therapeutic resistance. These findings caution against single-agent metabolic targeting and suggest PGK1 as a potential biomarker for TMZ response in GBM.
Collapse
Affiliation(s)
- Xing Li
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, Guangdong Province, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, 518055, Guangdong Province, China
| | - Wansong Zhang
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, Guangdong Province, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, 518055, Guangdong Province, China
| | - Yitong Fang
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, Guangdong Province, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, 518055, Guangdong Province, China
| | - Tianhu Sun
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, Guangdong Province, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, 518055, Guangdong Province, China
| | - Jian Chen
- Research Unit of Medical Neurobiology, Chinese Institute for Brain Research, Beijing, Chinese Academy of Medical Sciences, Beijing, China
| | - Ruilin Tian
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, Guangdong Province, China.
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, 518055, Guangdong Province, China.
| |
Collapse
|
34
|
Sui Y, Hoshi N, Okamoto N, Inoue Y, Funatsu T, Ku Y, Ooi M, Watanabe D, Miyazaki H, Agawa M, Nakamura H, Ohgaki R, Kanai Y, Yang H, Kodama Y. The role of LAT1 in AOM/DSS-induced colorectal tumorigenesis. Biochem Biophys Res Commun 2025; 751:151446. [PMID: 39922055 DOI: 10.1016/j.bbrc.2025.151446] [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: 11/14/2024] [Revised: 01/25/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
Amino acid transporters are essential for supplying nutrients to cells and are implicated in tumor progression. L-type amino acid transporter 1 (LAT1) is reported to be overexpressed in various cancers, affecting tumor development. However, the exact mechanisms by which LAT1 affects colorectal cancer (CRC) arising from a chronic inflammatory background are not yet fully understood. This study aimed to explore the role of LAT1 in CRC. Mice with intestinal epithelium-specific deletions of LAT1 (LAT1fl/fl; vil-cre) were treated with azoxymethane (AOM)/dextran sulfate sodium (DSS) in a colitis-associated cancer (CAC) model. Our results demonstrated that LAT1 was detected in normal colon crypts and highly expressed in AOM/DSS-induced tumor tissue. During the chronic colitis phase, weight loss was more prominent in LAT1fl/fl; vil-cre mice, compared with that in LAT1fl/fl mice. IL-1β and IL-6 expressions significantly increased in LAT1-deleted tumors; however, no overall difference in colon tumor number or size was observed between LAT1fl/fl and LAT1fl/fl; vil-cre mice. Accordingly, cell proliferation and apoptotic cell number were similar when comparing LAT1-deleted tumors with those with sufficient LAT1. Our findings indicated that LAT1 might not phenotypically affect overall colonic tumor development in this model; however, it affected the chronic colitis phase and inflammatory status within the tumors. These findings suggest that severe inflammation in tumors might have compensated for tumor growth in defects of amino acid supplementation through LAT1 deficiency, and provide insights into the potential of LAT1-targeted therapies for clinical CRC treatment.
Collapse
Affiliation(s)
- Yunlong Sui
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510260, China; Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Namiko Hoshi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan; Division of Integrated Analyses of Bioresource and Health Care, Kobe University Graduate School of Medicine, Hyogo, 650-0047, Japan.
| | - Norihiro Okamoto
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Yuta Inoue
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Takumi Funatsu
- Division of Integrated Analyses of Bioresource and Health Care, Kobe University Graduate School of Medicine, Hyogo, 650-0047, Japan
| | - Yuna Ku
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Makoto Ooi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Daisuke Watanabe
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Haruka Miyazaki
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Misaki Agawa
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Hirotaka Nakamura
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Ryuichi Ohgaki
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, 565-0871, Japan
| | - Yoshikatsu Kanai
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, 565-0871, Japan; Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Osaka, 565-0871, Japan
| | - Hui Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510260, China
| | - Yuzo Kodama
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| |
Collapse
|
35
|
Berens EB, Khou S, Huang E, Hoffman A, Johnson B, Kirchberger N, Sivagnanam S, Calistri NL, Derrick D, Liby TA, McLean IC, Alanizi AA, Ozmen F, Ozmen TY, Mills GB, Shelley Hwang E, Schedin PJ, Gonzalez H, Werb Z, Heiser LM, Coussens LM. Neoplastic immune mimicry potentiates breast tumor progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.17.633673. [PMID: 39896558 PMCID: PMC11785120 DOI: 10.1101/2025.01.17.633673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Dedifferentiation programs are commonly enacted during breast cancer progression to enhance tumor cell fitness. Increased cellular plasticity within the neoplastic compartment of tumors correlates with disease aggressiveness, often culminating in greater resistance to cytotoxic therapies or augmented metastatic potential. Here we report that subpopulations of dedifferentiated neoplastic breast epithelial cells express canonical leukocyte cell surface receptor proteins and have thus named this cellular program "immune mimicry." We document neoplastic cells engaging in immune mimicry within public human breast tumor single-cell RNA-seq datasets, histopathological breast tumor specimens, breast cancer cell lines, as well as in murine transgenic and cell line-derived mammary cancer models. Immune-mimicked neoplastic cells harbor hallmarks of dedifferentiation and are enriched in treatment-resistant and high-grade breast tumors. We corroborated these observations in aggressive breast cancer cell lines where anti-proliferative cytotoxic chemotherapies drove epithelial cells toward immune mimicry. Moreover, in subsequent proof-of-concept studies, we demonstrate that expression of the CD69 leukocyte activation protein by neoplastic cells confers a proliferative advantage that facilitates early tumor growth and therefore conclude that neoplastic breast epithelial cells upregulating leukocyte surface receptors potentiate malignancy. Moving forward, neoplastic immune mimicry should be evaluated for prognostic utility in breast cancer to determine stratification potential for patients with increased risks of tumor recurrence, metastasis, and therapeutic resistance. Statement of Significance Neoplastic breast epithelial cells express surface receptors canonically attributed to leukocytes and are associated with therapy resistance and aggressive tumor behavior.
Collapse
|
36
|
Ding J, Xie Y, Liu Z, Zhang Z, Ni B, Yan J, Zhou T, Hao J. Hypoxic and Acidic Tumor Microenvironment-Driven AVL9 Promotes Chemoresistance of Pancreatic Ductal Adenocarcinoma via the AVL9-IκBα-SKP1 Complex. Gastroenterology 2025; 168:539-555.e5. [PMID: 39566663 DOI: 10.1053/j.gastro.2024.10.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 09/24/2024] [Accepted: 10/22/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND & AIMS Gemcitabine combined with albumin-paclitaxel (AG) is a crucial therapeutic option for pancreatic ductal adenocarcinoma (PDAC). However, the response to chemotherapy is relatively poor, with rapid development of resistance. The aim of this study was to explore the mechanism of resistance to AG and to develop strategies that can sensitize the AG regimen. METHODS We used organoid models, patient-derived xenografts, and genetically engineered mouse models in our study. Chromatin immunoprecipitation, double luciferase assay, co-immunoprecipitation, and far-western blotting analysis were performed to investigate the mechanism. The AVL9 inhibitors were identified through protein structure analysis and molecular docking analysis, and their efficacy was verified in patient-derived xenografts, patient-derived organoids-based xenograft, and KPC models. RESULTS Through multistrategy screening, we identified AVL9 as a key target for AG resistance in PDAC. Its tumor-promoting effects were confirmed in our clinical cohorts. Mechanistically, HIF-1α, a hypoxia-related transcription factor, drives the expression of AVL9. AVL9 acts as a scaffold that facilitates the binding of IκBα to SKP1, leading to enhanced ubiquitination and degradation of IκBα, which further activates the nuclear factor-κB pathway. The potential AVL9-targeting inhibitor, Edotecarin, was shown to reverse AG chemo-resistance in PDAC. CONCLUSION AVL9 expression is driven by HIF-1α in PDAC. The physical interaction of AVL9, IκBα, and SKP1 provides a novel molecular mechanism for the abnormal activation of the nuclear factor-κB pathway. Therefore, the AVL9-targeting drug Edotecarin could be a promising therapeutic strategy for sensitizing PDAC to AG.
Collapse
Affiliation(s)
- Jinsheng Ding
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China; Department of Breast Oncoplastic Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, People's Republic of China
| | - Yongjie Xie
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Ziyun Liu
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Zhaoyu Zhang
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Bo Ni
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Jingrui Yan
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Tianxing Zhou
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China.
| | - Jihui Hao
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China.
| |
Collapse
|
37
|
Shan X, Cai Y, Zhu B, Sun X, Zhou L, Zhao Z, Li Y, Wang D. Computer-Aided Design of Self-Assembled Nanoparticles to Enhance Cancer Chemoimmunotherapy via Dual-Modulation Strategy. Adv Healthc Mater 2025; 14:e2404261. [PMID: 39828527 DOI: 10.1002/adhm.202404261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/05/2025] [Indexed: 01/22/2025]
Abstract
The rational design of self-assembled compounds is crucial for the highly efficient development of carrier-free nanomedicines. Herein, based on computer-aided strategies, important physicochemical properties are identified to guide the rational design of self-assembled compounds. Then, the pharmacophore hybridization strategy is used to design self-assemble nanoparticles by preparing new chemical structures by combining pharmacophore groups of different bioactive compounds. Hydroxychloroquine is grafted with the lipophilic vitamin E succinate and then co-assembled with bortezomib to fabricate the nanoparticle. The nanoparticle can reduce M2-type tumor-associated macrophages (TAMs) through lysosomal alkalization and induce immunogenic cell death (ICD) and nuclear factor-κB (NF-κB) inhibition in tumor cells. In mouse models, the nanoparticles induce decreased levels of M2-type TAMs, regulatory T cells, and transforming growth factor-β (TGF-β), and increase the proportion of cytotoxicity T lymphocytes. Additionally, the nanoparticles reduce the secretion of Interleukin-6 (IL-6) by inhibiting NF-κB and enhance the programmed death ligand-1 (PD-L1) checkpoint blockade therapy. The pharmacophore hybridization-derived nanoparticle provides a dual-modulation strategy to reprogram the tumor microenvironment, which will efficiently enhance the chemoimmunotherapy against triple-negative breast cancer.
Collapse
Affiliation(s)
- Xiaoting Shan
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Ying Cai
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Shandong, 264000, China
| | - Binyu Zhu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Xujie Sun
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Lingli Zhou
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Zhiwen Zhao
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Shandong, 264000, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264000, China
| | - Dangge Wang
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| |
Collapse
|
38
|
Wang W, Zhang S, Wang C, Gao S, Zhang L, Zhang C, Zheng Z, Zhang J, Xu H, Bo C, Li N. Fluorouracil enhances the anti-pancreatic cancer effect of anti-PD-L1 antibodies via up-regulating the expression of PD-L1 in cancer cells. J Investig Med 2025; 73:310-319. [PMID: 39760164 DOI: 10.1177/10815589251314192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Pancreatic cancer is characterized by occult onset, low early diagnosis rate, rapid progress, and poor prognosis. Due to the low response rate and low programmed cell death ligand 1 (PD-L1) expression in pancreatic cancer, the therapeutic application of PL-L1 inhibitors in pancreatic cancer is greatly limited. In vitro studies showed that the expression of PD-L1 increased in pancreatic cancer cells stimulated by fluorouracil (5-FU). We aim to explore the combined effect of 5-FU and anti-PD-L1 antibodies and to provide a reference for the clinical application of PD-L1 antibodies in pancreatic cancer. In the current study, male BALB/c mice were adopted to construct a tumor-bearing model of pancreatic cancer cells. 5-FU and anti-mouse PD-L1 antibodies were combined and administered to evaluate their synergistic effects. The enhancing immune cytotoxicity effect of 5-FU sensitizing the anti-PD-L1 antibody in vivo and in vitro was analyzed by immunohistochemistry and western blot assays. Results showed that 5-FU and anti-PD-L1 antibody combination increased the expression of PD-L1 and IFN-γ, and infiltration of CD8+ T lymphocytes in pancreatic xenograft tumor tissues, which was proven by immunohistochemistry and western analysis. Moreover, the combination with the 5-FU remarkably enhanced the immune cytotoxicity of anti-PD-L1 antibodies in mice. In vitro analysis demonstrates that 5-FU increases the expression of PD-L1 on the surface of pancreatic cancer cell lines via up-regulating nuclear factor kappa B (NF-κB) and Protein kinase B (AKT) pathways. This synergistic effect could be abolished by NF-κB and AKT inhibitors.
Collapse
Affiliation(s)
- Wei Wang
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Sujing Zhang
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Cong Wang
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Siming Gao
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Lingling Zhang
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Changwang Zhang
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Zheng Zheng
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Jiancong Zhang
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Hui Xu
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Changwen Bo
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Na Li
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| |
Collapse
|
39
|
Uthamacumaran A. Cell Fate Dynamics Reconstruction Identifies TPT1 and PTPRZ1 Feedback Loops as Master Regulators of Differentiation in Pediatric Glioblastoma-Immune Cell Networks. Interdiscip Sci 2025; 17:59-85. [PMID: 39420135 DOI: 10.1007/s12539-024-00657-4] [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: 10/11/2023] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 10/19/2024]
Abstract
Pediatric glioblastoma is a complex dynamical disease that is difficult to treat due to its multiple adaptive behaviors driven largely by phenotypic plasticity. Integrated data science and network theory pipelines offer novel approaches to studying glioblastoma cell fate dynamics, particularly phenotypic transitions over time. Here we used various single-cell trajectory inference algorithms to infer signaling dynamics regulating pediatric glioblastoma-immune cell networks. We identified GATA2, PTPRZ1, TPT1, MTRNR2L1/2, OLIG1/2, SOX11, FXYD6, SEZ6L, PDGFRA, EGFR, S100B, WNT, TNF α , and NF-kB as critical transition genes or signals regulating glioblastoma-immune network dynamics, revealing potential clinically relevant targets. Further, we reconstructed glioblastoma cell fate attractors and found complex bifurcation dynamics within glioblastoma phenotypic transitions, suggesting that a causal pattern may be driving glioblastoma evolution and cell fate decision-making. Together, our findings have implications for developing targeted therapies against glioblastoma, and the continued integration of quantitative approaches and artificial intelligence (AI) to understand pediatric glioblastoma tumor-immune interactions.
Collapse
Affiliation(s)
- Abicumaran Uthamacumaran
- Department of Physics (Alumni), Concordia University, Montréal, H4B 1R6, Canada.
- Department of Psychology (Alumni), Concordia University, Montréal, H4B 1R6, Canada.
- Oxford Immune Algorithmics, Reading, RG1 8EQ, UK.
| |
Collapse
|
40
|
Li Y, Miao J, Liu C, Tao J, Zhou S, Song X, Zou Y, Huang Y, Zhong L. Kushenol O Regulates GALNT7/NF-κB axis-Mediated Macrophage M2 Polarization and Efferocytosis in Papillary Thyroid Carcinoma. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156373. [PMID: 39864368 DOI: 10.1016/j.phymed.2025.156373] [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: 08/05/2024] [Revised: 12/17/2024] [Accepted: 01/02/2025] [Indexed: 01/28/2025]
Abstract
BACKGROUND The incidence of papillary thyroid carcinoma (PTC) is on the rise globally. It is frequently associated with early lymphatic metastasis, and the poor prognosis tends to be poor once metastasis or recurrence occurs, even with current treatment modalities. Kushenol O, a novel extract derived from Sophora flavescens, has shown remarkable anticancer properties. However, its specific role in the treatment of PTC remains to be elucidated. PURPOSE This objective of this study is to examine the effects of kushenol O on the proliferation and invasion capacity of PTC cells, as well as to delve into its potential mechanisms of action. METHODS Multi-omics was employed to identify the potential therapeutic targets for PTC. Single-cell RNA sequencing (scRNA-seq) investigated how these targets influence the remodeling of the tumor immune microenvironment (TIME). Kushenol O was employed to treat the PTC cell lines, with assessments conducted on its effects regarding cell viability, apoptosis, oxidative stress, and invasiveness. Molecular simulation was used to validate kushenol O's affinity for the therapeutic targets and biological toxicity. The impact of kushenol O was further evaluated using qRT-PCR and EdU assays, while cytotoxicity was measured by the CCK-8. RESULTS GALNT7 is a potential new target for the treatment of PTC. It may regulate the macrophage M2 polarization and efferocytosis in the TIME of PTC through regulating the NF-κB axis. Kushenol O inhibits PTC cells proliferation and promotes apoptosis by inhibiting the expression of GALNT7, induces a decrease in SOD levels and an increase in MDA levels by inhibiting mitochondrial function, and promotes the accumulation of ROS, which inhibits G1 phase and promotes early apoptosis. CONCLUSIONS Kushenol O may inhibit the inflammation-cancer transformation and tumor progression of PTC by inhibiting GALNT7 and thus regulating NF-κB axis. These findings highlight the potential of kushenol O as immunomodulator or therapeutic agent, which may have important clinical implications.
Collapse
Affiliation(s)
- Yutong Li
- The First School of Clinical Medicine, Guangdong Medical University, Zhanjiang, Guangdong Province, 524023, China; The Department of General Surgery, Zhongshan City People's Hospital, Zhongshan, Guangdong Province, 528400, China
| | - Jianhang Miao
- The Department of General Surgery, Zhongshan City People's Hospital, Zhongshan, Guangdong Province, 528400, China
| | - Chizhuai Liu
- The Department of General Surgery, Zhongshan City People's Hospital, Zhongshan, Guangdong Province, 528400, China
| | - Jiahua Tao
- The First School of Clinical Medicine, Guangdong Medical University, Zhanjiang, Guangdong Province, 524023, China
| | - Sifan Zhou
- The First School of Clinical Medicine, Guangdong Medical University, Zhanjiang, Guangdong Province, 524023, China
| | - Xingyu Song
- The First School of Clinical Medicine, Guangdong Medical University, Zhanjiang, Guangdong Province, 524023, China
| | - Yecheng Zou
- The First School of Clinical Medicine, Guangdong Medical University, Zhanjiang, Guangdong Province, 524023, China
| | - Yuyang Huang
- The First School of Clinical Medicine, Guangdong Medical University, Zhanjiang, Guangdong Province, 524023, China
| | - Linkun Zhong
- The Department of General Surgery, Zhongshan City People's Hospital, Zhongshan, Guangdong Province, 528400, China.
| |
Collapse
|
41
|
Mirchandani AS, Sanchez-Garcia MA, Walmsley SR. How oxygenation shapes immune responses: emerging roles for physioxia and pathological hypoxia. Nat Rev Immunol 2025; 25:161-177. [PMID: 39349943 DOI: 10.1038/s41577-024-01087-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2024] [Indexed: 03/04/2025]
Abstract
Most eukaryotes require oxygen for their survival and, with increasing multicellular complexity, oxygen availability and delivery rates vary across the tissues of complex organisms. In humans, healthy tissues have markedly different oxygen gradients, ranging from the hypoxic environment of the bone marrow (where our haematopoietic stem cells reside) to the lungs and their alveoli, which are among the most oxygenated areas of the body. Immune cells are therefore required to adapt to varying oxygen availability as they move from the bone marrow to peripheral organs to mediate their effector functions. These changing oxygen gradients are exaggerated during inflammation, where oxygenation is often depleted owing to alterations in tissue perfusion and increased cellular activity. As such, it is important to consider the effects of oxygenation on shaping the immune response during tissue homeostasis and disease conditions. In this Review, we address the relevance of both physiological oxygenation (physioxia) and disease-associated hypoxia (where cellular oxygen demand outstrips supply) for immune cell functions, discussing the relevance of hypoxia for immune responses in the settings of tissue homeostasis, inflammation, infection, cancer and disease immunotherapy.
Collapse
Affiliation(s)
- Ananda Shanti Mirchandani
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
| | | | - Sarah Ruth Walmsley
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
42
|
Khalid AQ, Zaidan TN, Bhuvanendran S, Magalingam KB, Mohamedahmed SM, Ramdas P, Radhakrishnan AK. Insights into the Anticancer Mechanisms Modulated by Gamma and Delta Tocotrienols in Colorectal Cancers. Nutr Rev 2025; 83:e1295-e1310. [PMID: 39181121 PMCID: PMC11819494 DOI: 10.1093/nutrit/nuae108] [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] [Indexed: 08/27/2024] Open
Abstract
Colorectal cancer (CRC) is a growing concern all over the world. There has been a concerted effort to identify natural bioactive compounds that can be used to prevent or overcome this condition. Tocotrienols (T3s) are a naturally occurring form of vitamin E known for various therapeutic effects, such as anticancer, antioxidant, neuroprotective, and anti-inflammatory activities. The literature evidence suggests that two T3 analogues, ie, gamma (γ)- and delta (δ)-T3, can modulate cancers via several cancer-related signaling pathways. The aim of this review was to compile and analyze the existing literature on the diverse anticancer mechanisms of γT3 and δT3 exhibited in CRC cells, to showcase the anticancer potential of T3s. Medline was searched for research articles on anticancer effects of γT3 and δT3 in CRC published in the past 2 decades. A total of 38 articles (26 cell-based, 9 animal studies, 2 randomized clinical trials, and 1 scoping review) that report anticancer effects of γT3 and δT3 in CRC were identified. The findings reported in those articles indicate that γT3 and δT3 inhibit the proliferation of CRC cells, induce cell cycle arrest and apoptosis, suppress metastasis, and produce synergistic anticancer effects when combined with well-established anticancer agents. There is preliminary evidence that shows that T3s affect telomerase functions and support anticancer immune responses. γT3 and δT3 have the potential for development as anticancer agents.
Collapse
Affiliation(s)
- Ali Qusay Khalid
- Food as Medicine Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Malaysia
| | - Tabarek Najeeb Zaidan
- Department of Food Science and Nutrition, Faculty of Applied Sciences, UCSI University, UCSI Heights, Cheras, 56000 Kuala Lumpur, Malaysia
| | - Saatheeyavaane Bhuvanendran
- Food as Medicine Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Malaysia
| | - Kasthuri B Magalingam
- Food as Medicine Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Malaysia
| | - Shaza M Mohamedahmed
- Food as Medicine Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Malaysia
| | - Premdass Ramdas
- Food as Medicine Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Malaysia
| | - Ammu K Radhakrishnan
- Food as Medicine Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Malaysia
| |
Collapse
|
43
|
Romaniello D, Dall'Olio L, Mazzeschi M, Francia A, Pagano F, Gelfo V, D'Uva G, Giampieri E, Lauriola M. NF-kB oscillation profiles decode response to anti-EGFR monoclonal antibodies. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2025; 31:100219. [PMID: 39892619 DOI: 10.1016/j.slasd.2025.100219] [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: 09/28/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 02/04/2025]
Abstract
A direct connection between an inflammatory environment and cancer has been extensively proven over the years. We previously reported that the presence of interleukin 1 (IL-1) is responsible for the lack of response to monoclonal antibody targeting epidermal growth factor receptor (EGFR) in colorectal cancer (CRC). Considering the driver role of NF-kB in controlling the expression of IL-1, herein, we investigate the dynamics of the oscillatory profile of the NF-kB response to monoclonal antibody, on the background of an inflammatory environment. NF-kB is a typical transcription factor that displays intrinsic oscillatory behavior, whose biological relevance in term for example of decoding response to monoclonal antibodies, remains unclear. Using live cell luciferase techniques, we recorded NF-kB activity over time in response to cetuximab (CTX) alone or in combination with IL-1 cytokines. Our results revealed an additive effect of these two agents on NF-kB activation, which was specific to CTX responsive cells. In contrast, CTX resistant cells did not display a significant change in the NF-kB profile under the IL-1 plus CTX combination. These results suggest an immediate interactive crosstalk between IL-1 and EGFR in the activation of NF-kB signaling pathway, which may lay the basis for the development of drug tolerant persister cells (DTP), leading to CTX resistance.
Collapse
Affiliation(s)
- Donatella Romaniello
- Department of Medical and Surgical Sciences, University of Bologna, via Massarenti 9, 40138 Bologna, Italy
| | - Lorenzo Dall'Olio
- Laboratory of Data Science and Bioinformatics, IRCSS Institute of Neurological Sciences, Bellaria Hospital, via Altura 3, 40139 Bologna, Italy
| | - Martina Mazzeschi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, via Massarenti 9, 40138 Bologna, Italy
| | - Anna Francia
- Department of Medical and Surgical Sciences, University of Bologna, via Massarenti 9, 40138 Bologna, Italy
| | - Federica Pagano
- Department of Medical and Surgical Sciences, University of Bologna, via Massarenti 9, 40138 Bologna, Italy
| | - Valerio Gelfo
- Department of Medical and Surgical Sciences, University of Bologna, via Massarenti 9, 40138 Bologna, Italy
| | - Gabriele D'Uva
- Department of Medical and Surgical Sciences, University of Bologna, via Massarenti 9, 40138 Bologna, Italy; IRCCS Azienda Ospedaliero-Universitaria di Bologna, via Massarenti 9, 40138 Bologna, Italy
| | - Enrico Giampieri
- Department of Medical and Surgical Sciences, University of Bologna, via Massarenti 9, 40138 Bologna, Italy
| | - Mattia Lauriola
- Department of Medical and Surgical Sciences, University of Bologna, via Massarenti 9, 40138 Bologna, Italy; IRCCS Azienda Ospedaliero-Universitaria di Bologna, via Massarenti 9, 40138 Bologna, Italy.
| |
Collapse
|
44
|
Tripathi S, Sharma Y, Kumar D. Unveiling the link between chronic inflammation and cancer. Metabol Open 2025; 25:100347. [PMID: 39876904 PMCID: PMC11772974 DOI: 10.1016/j.metop.2025.100347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/05/2025] [Accepted: 01/06/2025] [Indexed: 01/31/2025] Open
Abstract
The highly nuanced transition from an inflammatory process to tumorigenesis is of great scientific interest. While it is well known that environmental stimuli can cause inflammation, less is known about the oncogenic modifications that chronic inflammation in the tissue microenvironment can bring about, as well as how these modifications can set off pro-tumorigenic processes. It is clear that no matter where the environmental factors come from, maintaining an inflammatory microenvironment encourages carcinogenesis. In addition to encouraging angiogenesis and metastatic processes, sustaining the survival and proliferation of malignant transformed cells, and possibly altering the efficacy of therapeutic agents, inflammation can negatively regulate the antitumoral adaptive and innate immune responses. Because chronic inflammation has multiple pathways involved in tumorigenesis and metastasis, it has gained recognition as a marker of cancer and a desirable target for cancer therapy. Recent advances in our knowledge of the molecular mechanisms that drive cancer's progression demonstrate that inflammation promotes tumorigenesis and metastasis while suppressing anti-tumor immunity. In many solid tumor types, including breast, lung, and liver cancer, inflammation stimulates the activation of oncogenes and impairs the body's defenses against the tumor. Additionally, it alters the microenvironment of the tumor. As a tactical approach to cancer treatment, these findings have underscored the importance of targeting inflammatory pathways. This review highlights the role of inflammation in cancer development and metastasis, focusing on its impact on tumor progression, immune suppression, and therapy resistance. It examines current anti-inflammatory strategies, including NSAIDs, cytokine modulators, and STAT3 inhibitors, while addressing their potential and limitations. The review emphasizes the need for further research to unravel the complex mechanisms linking inflammation to cancer progression and identify molecular targets for specific cancer subtypes.
Collapse
Affiliation(s)
- Siddhant Tripathi
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
| | - Yashika Sharma
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
| | - Dileep Kumar
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| |
Collapse
|
45
|
Caramella-Pereira F, Zheng Q, Hicks JL, Roy S, Jones T, Pomper M, Antony L, Meeker AK, Yegnasubramanian S, De Marzo AM, Brennen WN. Overexpression of fibroblast activation protein (FAP) in the stroma of proliferative inflammatory atrophy (PIA) and primary adenocarcinoma of the prostate. Pathology 2025:S0031-3025(25)00093-5. [PMID: 40187966 DOI: 10.1016/j.pathol.2024.12.637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 11/05/2024] [Accepted: 12/16/2024] [Indexed: 04/07/2025]
Abstract
Fibroblast activation protein (FAP) is a serine protease upregulated at sites of tissue remodelling and cancer that represents a promising therapeutic and molecular imaging target. In prostate cancer, studies of FAP expression using tissue microarrays are conflicting, such that its clinical potential is unclear. Furthermore, little is known regarding FAP expression in benign prostatic tissues. Here we demonstrated, using a novel iterative multiplex immunohistochemistry assay in standard tissue sections, that FAP was nearly absent in normal regions but was increased consistently in regions of proliferative inflammatory atrophy (PIA). In carcinoma, FAP was expressed in all cases but was highly heterogeneous. High FAP levels were associated with increased pathological stage and cribriform morphology. We verified that FAP levels in cancer correlated with CD163+ M2 macrophage density. In this first report to quantify FAP protein in benign prostate and primary tumours, using standard large tissue sections, we clarify that FAP is present in all primary prostatic carcinomas, supporting its potential clinical relevance. The finding of high levels of FAP within PIA supports the injury/regeneration model for its pathogenesis and suggests that it harbours a protumourigenic stroma, yet high levels of FAP in benign regions could lead to false-positive FAP-based molecular imaging results in clinically localised prostate cancer.
Collapse
Affiliation(s)
| | - Qizhi Zheng
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jessica L Hicks
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sujayita Roy
- Microbiology Devices for Regulatory Authorization or Clearance, Food and Drug Administration, Silver Spring, MD, USA
| | - Tracy Jones
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Martin Pomper
- Department of Radiology, UT Southwestern, Dallas TX, USA
| | - Lizamma Antony
- Department of Oncology, and Urology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins and the James Buchanan Brady Urological Research Institute, Baltimore, MD, USA
| | - Alan K Meeker
- Department of Pathology, Oncology, and Urology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins and the James Buchanan Brady Urological Research Institute, Baltimore, MD, USA
| | - Srinivasan Yegnasubramanian
- Departments of Oncology, Pathology and Radiation Oncology and Molecular Radiation Sciences, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins and the James Buchanan Brady Urological Research Institute, Baltimore, MD, USA
| | - Angelo M De Marzo
- Department of Pathology, Oncology, and Urology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins and the James Buchanan Brady Urological Research Institute, Baltimore, MD, USA.
| | - W Nathaniel Brennen
- Department of Oncology, and Urology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins and the James Buchanan Brady Urological Research Institute, Baltimore, MD, USA
| |
Collapse
|
46
|
Xu Y, Kang K, Coakley BA, Eisenstein S, Parveen A, Mai S, Wang YS, Zheng J, Boral D, Mai J, Pan W, Zhang L, Aaronson SA, Fang B, Divino C, Zhang B, Song WM, Hung MC, Pan PY, Chen SH. Modulation of tumor inflammatory signaling and drug sensitivity by CMTM4. EMBO J 2025; 44:1866-1883. [PMID: 39948411 PMCID: PMC11914105 DOI: 10.1038/s44318-024-00330-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 03/19/2025] Open
Abstract
Although inflammation has been widely associated with cancer development, how it affects the outcomes of immunotherapy and chemotherapy remains incompletely understood. Here, we show that CKLF-like MARVEL transmembrane domain-containing member 4 (CMTM4) is highly expressed in multiple human and murine cancer types including Lewis lung carcinoma, triple-negative mammary cancer and melanoma. In lung carcinoma, loss of CMTM4 significantly reduces tumor growth and impairs NF-κB, mTOR, and PI3K/Akt pathway activation. Furthermore, we demonstrate that CMTM4 can regulate epidermal growth factor (EGF) signaling post-translationally by promoting EGFR recycling and preventing its Rab-dependent degradation. Consequently, CMTM4 knockout sensitizes human lung tumor cells to EGFR inhibitors. In addition, CMTM4 knockout tumors stimulated with EGF show a decreased ability to produce inflammatory cytokines including granulocyte colony-stimulating factor (G-CSF), leading to decreased recruitment of polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) and therefore establishing a less suppressive tumor immune environment in both lung and mammary cancers. We also present evidence indicating that CMTM4-targeting siRNA-loaded liposomes reduce lung tumor growth in vivo and prolong animal survival. Knockout of CMTM4 enhances immune checkpoint blockade or chemotherapy to further reduce lung tumor growth. These data suggest that CMTM4 represents a novel target for the inhibition of tumor inflammation, and improvement of the immune response and tumor drug sensitivity.
Collapse
Affiliation(s)
- Yitian Xu
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Neal Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Kyeongah Kang
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Neal Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Brian A Coakley
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Samuel Eisenstein
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Arshiya Parveen
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Neal Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Sunny Mai
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Neal Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Yuan Shuo Wang
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Junjun Zheng
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Neal Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Debasish Boral
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Junhua Mai
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - William Pan
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Licheng Zhang
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Neal Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Stuart A Aaronson
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bingliang Fang
- Department of Thoracic and Cardiovascular Surgery, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Celia Divino
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Won-Min Song
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
| | - Ping-Ying Pan
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Shu-Hsia Chen
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA.
- Neal Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, 77030, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Physiology, Biophysics, and Systems Biology, Weill Cornell Medical Science and Graduate School of Medical Sciences, New York, NY, 10065, USA.
- Graduate and professional school at Texas A&M University, 400 Bizzell St., College Station, TX, 77840, USA.
| |
Collapse
|
47
|
Wang Y, Zhao D, Nong X. Artesunate alleviates radiation-induced submandibular gland epithelial cell damage in rats by reducing inflammation and apoptosis. Cell Biol Int 2025; 49:250-261. [PMID: 39607036 DOI: 10.1002/cbin.12261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/09/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024]
Abstract
Salivary hypofunction is a common complication in patients with head and neck cancers following radiotherapy (RT). RT-induced inflammation in salivary gland cells leads to apoptosis and fibrosis. Artesunate (ART) is a bioactive compound with anti-inflammatory and anti-fibrosis properties. This study aimed to investigate the protective effects of ART on X-ray-induced injury of submandibular gland (SMG) epithelial cells in rats. Second-generation SMG epithelial cells were randomly divided into five groups: natural control group (NC), irradiated group (IR), and irradiated groups treated with ART at concentrations of 5, 10, and 20 μM. Cells were harvested 48 h postirradiation for analysis. The results demonstrated that ART attenuated the damage to AQP5, a crucial indicator of salivary gland function, as evidenced by the decreased expression of AQP5 at both mRNA and protein levels. Additionally, ART decreased the expression of inflammatory cytokines: IL-6 and TNF-α. TUNEL staining revealed reduced apoptosis in the ART groups, particularly the IR + 10 μM group. RT-PCR and Western blot analysis of apoptosis cytokines Bax/Bcl-2 and Caspase-3 confirmed these findings. Furthermore, ART inhibited the expression of NF-κB at both mRNA and protein levels. In conclusion, these results suggest that ART may reduce inflammation and apoptosis in SMG epithelial cells following radiation by inhibiting the NF-κB pathway.
Collapse
Affiliation(s)
- Yuchen Wang
- College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Danni Zhao
- College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaolin Nong
- College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, China
| |
Collapse
|
48
|
Colella F, Calapà F, Artemi G, Pazzaglia E, Colonna R, Vitale S, Lazzarino G, Vincenzoni F, Fiori ME, De Maria R, Lucchisani S, Genovese G, Perelli L, Tavazzi B, Sgambato A, Lucchetti D. CD147 Mediates the Metabolic Reprogramming of Cancer Associated Fibroblasts Induced by EVs Released by Differentiating Cancer Stem Cells. JOURNAL OF EXTRACELLULAR BIOLOGY 2025; 4:e70039. [PMID: 40104175 PMCID: PMC11913892 DOI: 10.1002/jex2.70039] [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: 08/19/2024] [Revised: 01/24/2025] [Accepted: 01/31/2025] [Indexed: 03/20/2025]
Abstract
Several reports have demonstrated that CD147, an N-glycosylated protein that is exchanged by cells in soluble form or through small extracellular vesicles (sEVs), can promote cancer progression. However, its activity related to EVs in colorectal cancer (CRC) is still not fully understood. Previously, we showed that sEV secretion during CRC stem cell (CR-CSCs) differentiation is partially controlled by CD147, and that CD147-expressing sEVs (sEVs-CD147) activate a signalling cascade in recipient cells, inducing molecular invasive features in CR-CSCs. In the present study, we demonstrated that sEVs-CD147 increase the expression of myofibroblast and activation markers in cancer-associated fibroblasts (CAF). In sEVs-CD147-activated CAF, aerobic glycolysis was also triggered by the β-catenin signalling pathway and induced lactate release. These effects were associated with NFKβ upregulation and NO secretion that caused increased cytokines production and VEGF release, respectively. Furthermore, co-culture with CAF promoted CR-CSC invasivity in vitro and tumour growth in vivo. Spatial proteomics analysis confirmed in vivo the activation of fibroblasts and the modulation of their metabolic features, within their biological context, after their conditioning with CD147-expressing sEVs. Our findings indicate that sEV-packaged CD147 is involved in CAF activation, thus promoting tumour progression via stroma metabolism modification.
Collapse
Affiliation(s)
- Filomena Colella
- Multiplex Spatial Profiling Facility, Fondazione Policlinico Universitario 'Agostino Gemelli' IRCCS Rome Italy
| | - Federica Calapà
- Department of Oncology and Molecular Medicine Istituto Superiore di Sanità Rome Italy
| | - Giulia Artemi
- Department of Translational Medicine and Surgery Università Cattolica del Sacro Cuore Rome Italy
| | - Erica Pazzaglia
- Department of Translational Medicine and Surgery Università Cattolica del Sacro Cuore Rome Italy
| | - Rita Colonna
- Department of Translational Medicine and Surgery Università Cattolica del Sacro Cuore Rome Italy
| | - Sara Vitale
- Department of Oncology and Molecular Medicine Istituto Superiore di Sanità Rome Italy
| | - Giacomo Lazzarino
- UniCamillus - Saint Camillus International University of Health and Medical Sciences Rome Italy
| | - Federica Vincenzoni
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie Università Cattolica del Sacro Cuore Rome Italy
| | - Micol Eleonora Fiori
- Department of Oncology and Molecular Medicine Istituto Superiore di Sanità Rome Italy
| | - Ruggero De Maria
- Multiplex Spatial Profiling Facility, Fondazione Policlinico Universitario 'Agostino Gemelli' IRCCS Rome Italy
- Department of Translational Medicine and Surgery Università Cattolica del Sacro Cuore Rome Italy
| | - Sara Lucchisani
- Department of Oncology and Molecular Medicine Istituto Superiore di Sanità Rome Italy
| | - Giannicola Genovese
- Department of Genitourinary Medical Oncology The University of Texas MD Anderson Cancer Center Houston USA
| | - Luigi Perelli
- Department of Genitourinary Medical Oncology The University of Texas MD Anderson Cancer Center Houston USA
| | - Barbara Tavazzi
- UniCamillus - Saint Camillus International University of Health and Medical Sciences Rome Italy
| | - Alessandro Sgambato
- Multiplex Spatial Profiling Facility, Fondazione Policlinico Universitario 'Agostino Gemelli' IRCCS Rome Italy
- Department of Translational Medicine and Surgery Università Cattolica del Sacro Cuore Rome Italy
| | - Donatella Lucchetti
- Multiplex Spatial Profiling Facility, Fondazione Policlinico Universitario 'Agostino Gemelli' IRCCS Rome Italy
- Department of Translational Medicine and Surgery Università Cattolica del Sacro Cuore Rome Italy
| |
Collapse
|
49
|
You YL, Byun HJ, Chang YB, Kim H, Lee H, Suh HJ, Jeon JY, Kim BR, Hwang JE, Lee JH, Choi HS. Euglena gracilis-derived β-glucan ameliorates particulate matter (PM 2.5)-induced airway inflammation by modulating nuclear factor kappa B, mitogen-activated protein kinase, and nuclear factor erythroid 2-related factor 2 signaling pathways in A549 cells and BALB/c mice. Int J Biol Macromol 2025; 296:139671. [PMID: 39798741 DOI: 10.1016/j.ijbiomac.2025.139671] [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: 09/29/2024] [Revised: 12/19/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
This study aimed to investigate the effects of β-glucan derived from Euglena gracilis (EGB), an edible microalga, on particulate matter (PM2.5)-induced airway inflammation in A549 cells and BALB/c mice. EGB effectively suppressed the mRNA and protein levels of inflammatory cytokines (IL-6, IL-1β, TNF-α, IL-8) and mediators (iNOS, COX-2), while inhibiting the NF-κB and MAPK signaling pathways triggered by PM2.5 exposure and reducing nuclear NF-κB levels. Additionally, EGB decreased PM2.5-induced ROS production and increased the protein levels of NRF2 and HO-1, along with genes encoding antioxidant enzymes (catalase, GPx, SOD1), associated with elevated nuclear NRF2 levels. EGB reduced immune cell infiltration and inflammatory cytokine levels in BALF and serum, both of which increased by PM2.5 exposure. EGB also significantly increased alveolar numbers while decreasing the gene expression of MMP1/9/13. Furthermore, EGB suppressed PM2.5-induced bronchial thickening and collagen-1 deposition by downregulating TGF-β1 expression, and alleviated goblet cell hyperplasia and mucin production in lung tissues. These results suggest that EGB effectively reduces PM2.5-induced airway inflammation by suppressing NF-κB and MAPK signaling pathways, lowering pro-inflammatory cytokines, and activating the NRF2-HO-1 signaling pathway to enhance antioxidant enzyme expression. This study highlights the potential of EGB as an edible functional agent for controlling PM-related airway inflammation.
Collapse
Affiliation(s)
- Ye-Lim You
- Department of Food Nutrition, Sangmyung University, Seoul 03016, Republic of Korea
| | - Ha-Jun Byun
- Department of Food Nutrition, Sangmyung University, Seoul 03016, Republic of Korea
| | - Yeok Boo Chang
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Hyeongyeong Kim
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Hyowon Lee
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Hyung Joo Suh
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Jin-Young Jeon
- BlueBIO CIC, Daesang Corp., Seoul 07789, Republic of Korea
| | - Bo-Ra Kim
- BlueBIO CIC, Daesang Corp., Seoul 07789, Republic of Korea
| | - Ji Eun Hwang
- BlueBIO CIC, Daesang Corp., Seoul 07789, Republic of Korea
| | - Jun Hee Lee
- Health R&D Institute, Daesang Corp., Seoul 07789, Republic of Korea
| | - Hyeon-Son Choi
- Department of Food Nutrition, Sangmyung University, Seoul 03016, Republic of Korea.
| |
Collapse
|
50
|
Hu CH, Chen Y, Jin TY, Wang Z, Jin B, Liao J, Ding CY, Zhang A, Tang WY, Zhang LX, Xu LY, Ning FM, Liang G, Wei XH, Wang Y. A derivative of tanshinone IIA and salviadione, 15a, inhibits inflammation and alleviates DSS-induced colitis in mice by direct binding and inhibition of RIPK2. Acta Pharmacol Sin 2025; 46:672-686. [PMID: 39443729 PMCID: PMC11845706 DOI: 10.1038/s41401-024-01399-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024]
Abstract
Inflammatory bowel diseases (IBDs) are chronic inflammatory conditions primarily affecting the gastrointestinal tract. Previous studies established the role of the NF-κB signaling pathway in the development of IBDs, suggesting that anti-inflammatory therapies might offer a viable treatment strategy. Tanshinone IIA and salviadione, both derived from Salviae Miltiorrhizae Radix et Rhizoma, possess anti-inflammatory and anti-oxidative activities. A series of new compounds were synthesized by hybridizing salviadione with tanshinone. Among these compounds, 15a showed beneficial effects in LPS-induced acute lung injury and diabetes-induced renal injury mouse models. The current study explored the therapeutic efficacy of 15a using both acute and chronic colitis models and elucidated the underlying mechanisms. DSS-induced colitis models were established in mice, where acute colitis was treated with compound 15a (5 or 10 mg·kg-1·d-1) for 8 days, while chronic colitis mice received compound 15a (5 or 10 mg·kg-1·d-1, i.g.) during 2.5% DSS administration. The 15a treatment significantly alleviated DSS-induced pathological and inflammatory damages in both acute and chronic colitis mouse models. In mouse intestinal epithelial cell line MODE-K, pretreatment with compound 15a (5 or 10 μM) significantly suppressed LPS + L18-MDP-induced inflammatory responses. The receptor-interacting serine/threonine kinase 2 (RIPK2) was identified as a direct binding target of compound 15a using microarrays and recombinant human proteins. Moreover, 15a could directly bind to and inhibit the phosphorylation of RIPK2, leading to the suppression of the NF-κB and MAPK signaling pathways. Furthermore, LEU153 and VAL32 were identified within the KD domain of RIPK2 as critical amino residues for the binding of 15a. Briefly, the current findings demonstrate that compound 15a holds promise as a therapeutic agent for managing acute and chronic colitis.
Collapse
Affiliation(s)
- Cheng-Hong Hu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Yue Chen
- Chemical Biology Research Center, Wenzhou Medical University, School of Pharmaceutical Sciences, Wenzhou, 325035, China
| | - Tian-Yang Jin
- Chemical Biology Research Center, Wenzhou Medical University, School of Pharmaceutical Sciences, Wenzhou, 325035, China
| | - Zhe Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Bo Jin
- Chemical Biology Research Center, Wenzhou Medical University, School of Pharmaceutical Sciences, Wenzhou, 325035, China
| | - Jing Liao
- Chemical Biology Research Center, Wenzhou Medical University, School of Pharmaceutical Sciences, Wenzhou, 325035, China
| | - Chun-Yong Ding
- Pharm-X Center, College of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ao Zhang
- Pharm-X Center, College of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wei-Yang Tang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Ling-Xi Zhang
- Chemical Biology Research Center, Wenzhou Medical University, School of Pharmaceutical Sciences, Wenzhou, 325035, China
| | - Lei-Yu Xu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Fang-Min Ning
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Guang Liang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 310051, China
| | - Xiao-Hong Wei
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China.
| | - Yi Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China.
| |
Collapse
|