1
|
Kim HD, Choi H, Park JY, Kim CH. Distinct structural basis and catalytic classification of matrix metalloproteinases and their endogenous tissue inhibitors with glycosylation issue in cellular and tissue regulation. Arch Biochem Biophys 2025; 769:110436. [PMID: 40280381 DOI: 10.1016/j.abb.2025.110436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Revised: 04/22/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Matrix metalloproteinase (MMP) enzymes cleave proteins on the extracellular matrix (ECM) region. MMPs are categorized as Zn2+-binding endo-proteinases. MMPs are stringently regulated in cancers, inflammatory cells and tissues. There are 29 types of MMPs as initially expressed in inactive zymogens (proMMPs) and activated by proteolysis in vertebrates including human. MMPs consist of three highly conserved parts of pro-MMP in precursor, catalytic and hemopexin domains. The MMPs are composed of systemic complexes with their endogenously expressed inhibitors of the tissue inhibitors of metalloproteinases (TIMPs). Therefore, TIMPs intrinsically control such activated MMPs, indicating the existence of self-modulation capacity. N-linked glycosylation (N-glycosylation) saves biological information than known phosphorylation, ubiquitination and acetylation. The MMPs are roughly present as membrane-merged and secreted glycoproteins. MMPs N-glycans regulate cellular behaviors, immune tolerance, and developing angiogenesis. Aberrant N-glycosylation of MMPs may cause the pathogenic properties. N-glycosylation shapes phenotypes of MMPs-producing cells during early MMPs involved in human. Additionally, issues of MMPs and TIMPs glycosylation have been described to view the importance of the glycans in their interaction with owns and other targets. Most of MMPs and 4 TIMPs are not well studied for their glycosylation and its functional roles.
Collapse
Affiliation(s)
- Hee-Do Kim
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Suwon, Gyunggi-Do, 16419, Republic of Korea
| | - Hyunju Choi
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Suwon, Gyunggi-Do, 16419, Republic of Korea
| | - Jun-Young Park
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Suwon, Gyunggi-Do, 16419, Republic of Korea; Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon, 34141, Republic of Korea
| | - Cheorl-Ho Kim
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Suwon, Gyunggi-Do, 16419, Republic of Korea; Samsung Advanced Institute of Health Science and Technology (SAIHST), Sungkyunkwan University, Seoul, 06351, Republic of Korea.
| |
Collapse
|
2
|
Luo W, Sun Y, Cao L. TSPAN31 Activates Fatty Acid Metabolism and PI3K/AKT Pathway to Promote Tumor Progression in Breast Cancer. Mol Carcinog 2025; 64:1078-1089. [PMID: 40135650 DOI: 10.1002/mc.23912] [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: 12/11/2024] [Revised: 03/04/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025]
Abstract
Breast cancer (BC) is one of the most common human malignancies, but the mechanisms of BC have not been fully elucidated. Recently, tetraspanin 31 (TSPAN31) is reported to be linked to cancer progression. However, the function of TSPAN31 remains unclear in BC. Investigation of the function and potential mechanism of TSPAN31 in BC was the purpose of this study. Immunohistochemistry, western blot, and quantitative real-time polymerase chain reaction were applied to measure TSPAN31 expression. Loss and gain functional experiments were utilized to survey the influences of TSPAN31 on BC biological process, including cell growth, invasion, migration, and fatty acid metabolism. Mechanistically, Kyoto Encyclopedia of Genes and Genomes analysis based on DepMap database and Gene Set Enrichment Analysis based on The Cancer Genome Atlas database were executed to find TSPAN31-related pathway. Western blot was carried out to assess the changes of fatty acid synthase (FASN), sterol regulatory element binding protein 1 (SREBP1), acyl-CoA synthetase long-chain family member 1 (ACSL1), phosphatidylinositol 3-kinase (PI3K), phosphorylated (p)-PI3K, protein kinase B (AKT), and p-AKT. In human non-triple negative breast cancer tissues and cells, TSPAN31 expression was upregulated. TSPAN31 knockdown induced BC cell apoptosis, inhibited cell proliferation, invasion, migration, and fatty acid metabolism, and reduced the protein levels of FASN, SREBP1, ACSL1, p-PI3K/PI3K, and p-AKT/AKT. In contrast, TSPAN31 overexpression led to the opposite results. Additionally, the activator of PI3K (740 Y-P) attenuated the inhibition of TSPAN31 knockdown on fatty acid metabolism, proliferation, and invasion in BC cells. Through activation of fatty acid metabolism and PI3K/AKT pathway, TSPAN31 played a carcinogenic role in BC. For the mechanism of BC tumorigenesis, our study provides an interesting insight.
Collapse
Affiliation(s)
- Wenquan Luo
- Breast and Thyroid Surgery Department, Feicheng People's Hospital, Feicheng, Shandong, China
| | - Yuxiang Sun
- Breast and Thyroid Surgery Department, Feicheng People's Hospital, Feicheng, Shandong, China
| | - Liang Cao
- Radiotherapy Department, Taian Tumor Prevention and Treatment Hospital, Taian, Shandong, China
| |
Collapse
|
3
|
Zhou Y, Li W, Chen Y, Hu X, Miao C. Research progress on the impact of opioids on the tumor immune microenvironment (Review). Mol Clin Oncol 2025; 22:53. [PMID: 40297497 PMCID: PMC12035512 DOI: 10.3892/mco.2025.2848] [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: 11/13/2024] [Accepted: 03/17/2025] [Indexed: 04/30/2025] Open
Abstract
Opioids have been extensively used in cancer pain management because they can significantly improve the quality of life of patients with advanced cancer. However, recent evidence suggests that opioids can also modulate the tumor immune microenvironment by interacting with opioid receptors on immune cells, potentially regulating tumor progression and efficacy of cancer treatments. Notably, morphine can exhibit a dose-dependent effect on tumor immunity in pancreatic cancer and renal cell models, with lower doses potentially promoting tumor migration and invasion of pancreatic cancer cells, whereas higher doses shows the effect of inhibiting migration and invasion through distinct molecular pathways. The present review therefore comprehensively explored the mechanisms by which opioids can regulate the tumor immune microenvironment, focusing on their effects on immune cells, oxidative stress and angiogenesis. It also examined the interactions between opioids and other analgesics, along with their potential impact on immune modulation. All relevant articles and materials were retrieved from PubMed using the key words 'opioids', 'immune system', 'T cells', 'monocytes', 'macrophages', 'lymphocytes', 'natural killer cell', 'immunotherapy', 'immune cell function' and 'dose dependent effect'. The immunosuppressive effects of opioids, particularly through the µ-opioid receptor, can suppress the activity of natural killer cells, impair antigen presentation and promote the function of regulatory T cells (Tregs). These effects may contribute to tumor progression and metastasis. The severity of these immunosuppressive effects appears to be dose-dependent and can vary among different tumor types. There is evidence to suggest that tumors with higher immune responsiveness will experience more pronounced suppression, including the reduction of tumor angiogenesis, resulting in a decrease in tumor volume and decrease in tumor metastases. Furthermore, the combination of opioids with other analgesics, such as non-steroidal anti-inflammatory drugs, has the potential to exacerbate immunosuppression, which can in turn increase the risk of infections. Therefore, although opioids are essential for pain management in patients with cancer, their potential to modulate the immune microenvironment and promote tumor progression requires careful consideration. Clinicians should evaluate the advantages and disadvantages of opioids, especially regarding emerging immunotherapies, to minimize their potential negative effects on the outcomes of cancer treatments. Future studies are recommended to prioritize the development of strategies that optimize pain management whilst preserving immune function, such as receptor-specific opioid formulations or adjunctive therapies targeting immunosuppressive pathways.
Collapse
Affiliation(s)
- Yuancheng Zhou
- Department of Preventive Medicine, (Institute of Radiation Medicine), Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 251016, P.R. China
| | - Wenyu Li
- The Second School of Clinical Medicine of Binzhou Medical University, Anesthesiology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Yuanji Chen
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Xudong Hu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Chuanwang Miao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
4
|
Haddad TS, Bokhorst JM, van den Dobbelsteen L, Öztürk SK, Baumann E, van Vliet S, Verrijp K, Jamieson NB, Wood C, Berger MD, Kirsch R, Aben M, Rutgers N, Ueno H, Ciompi F, Simmer F, van der Laak J, Lugli A, Zlobec I, Nagtegaal I. Tumor budding and poorly differentiated clusters as a biological continuum in colorectal cancer invasion and prognosis. Sci Rep 2025; 15:16944. [PMID: 40374662 DOI: 10.1038/s41598-025-00866-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 05/02/2025] [Indexed: 05/17/2025] Open
Abstract
Tumor budding (TB) and poorly differentiated clusters (PDCs) are features of infiltrative growth patterns and powerful independent prognostic factors in colorectal cancer (CRC), yet the underlying biological mechanisms behind their role in CRC invasion is less understood. The aim of this study was to investigate the molecular background and prognostic role of tumor cluster size at the invasive margin (IM) of CRC, and determine whether a biological continuum between TB and PDCs exists. Using a combination of spatial transcriptomic and immunohistochemical (IHC) techniques, we demonstrated a biological continuum from larger to smaller tumor clusters, with TB possessing greater invasive potential than PDCs. We deployed artificial intelligence on a cohort of 1134 Stage I-III CRC resections to automatically detect nearly 400,000 isolated tumor cells/clusters of any particular size across the IM. We determined that 2-celled clusters were the most abundant feature at the IM, and the simultaneous assessment of TB and PDCs yielded a prognostic performance stronger than either independently. Our study provides a deeper understanding of the mechanisms behind CRC invasion while improving risk stratification for Stage I-III CRC.
Collapse
Affiliation(s)
| | | | | | - Sonay K Öztürk
- Radboud University Medical Center, Nijmegen, Netherlands
| | - Elias Baumann
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | | | - Kiek Verrijp
- Radboud University Medical Center, Nijmegen, Netherlands
| | - Nigel B Jamieson
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Colin Wood
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Martin D Berger
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Marco Aben
- Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Hideki Ueno
- National Defense Medical College, Saitama, Japan
| | | | - Femke Simmer
- Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Alessandro Lugli
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Inti Zlobec
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Iris Nagtegaal
- Radboud University Medical Center, Nijmegen, Netherlands.
| |
Collapse
|
5
|
Ding Q, Aung PP, Sun H, Soto LMS, Lu W, Raso MG, Rizvi YQ, Wistuba II, Sahin AA, Prieto VG. Upregulation of MMP9 expression in postradiation angiosarcoma of the breast: a possible predictive risk factor. Virchows Arch 2025:10.1007/s00428-025-04116-3. [PMID: 40335788 DOI: 10.1007/s00428-025-04116-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/14/2025] [Accepted: 04/24/2025] [Indexed: 05/09/2025]
Abstract
Postradiation angiosarcoma of the breast is a secondary angiosarcoma in breast carcinoma patients who have undergone radiation therapy. Although postradiation angiosarcoma is rare, it is very aggressive and has an extremely poor prognosis. Previous studies have implicated matrix metalloproteinase 9 (MMP9) in some cancer development and progression and angiogenesis. In this study, we compared MMP9 expression in surgical specimens from breast cancer patients treated with radiation who did and did not develop breast angiosarcoma within 15 years. We also compared MMP expression in the breast angiosarcoma specimens from patients with postradiation angiosarcoma of the breast and patients with non-breast angiosarcoma not previously treated with radiation. Our results showed that MMP9 was expressed in the endothelial cells in the breast carcinoma specimens from patients who developed angiosarcoma after radiation therapy but not in the breast carcinoma specimens from patients without angiosarcoma. In addition, MMP9 expression was higher in patients with postradiation angiosarcoma of the breast than in patients with non-radiation-associated primary angiosarcomas at non-breast sites. These results suggest that MMP9 may be a risk factor for and play a significant role in the development of postradiation angiosarcoma of the breast.
Collapse
Affiliation(s)
- Qingqing Ding
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Phyu P Aung
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Hongxia Sun
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Luisa Maren Solis Soto
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Wei Lu
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Maria Gabriela Raso
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yasmeen Qamar Rizvi
- Department of Investigative Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Aysegul A Sahin
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Victor G Prieto
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
6
|
Bruschi A, Sambri A, Fiore M, Bubbico E, Scollo C, Pace A, Zunarelli R, Montanari A, Cappelli A, Di Prinzio L, De Paolis M. Inside a Metastatic Fracture: Molecular Bases and New Potential Therapeutic Targets. Cancer Med 2025; 14:e70901. [PMID: 40304052 PMCID: PMC12041892 DOI: 10.1002/cam4.70901] [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: 01/12/2025] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 05/02/2025] Open
Abstract
INTRODUCTION Bone metastases and pathological fractures significantly impact the prognosis and quality of life in cancer patients. However, clinical and radiological features alone have been shown to fail to predict skeletal related events of a bone metastasis (SREs). AIM This study focuses on key molecular players including Matrix Metalloproteinases (MMPs), Integrins, Bone Morphogenetic Proteins (BMPs), Parathormone-related Protein (PTHrP). RESULTS The RANK/RANKL/Osteoprotegerin (OPG) pathway, and N-terminal peptide (NTx), involved in the metastatic process and bone integrity disruption. Elevated levels of these molecules have been pointed out as potential biomarkers for predicting SREs, but they have been poorly investigated. Moreover, batimastat, marimastat, tanomastat, andecaliximab, and HIV protease targeting MMPs; Volociximab/M200, cilengitide, abituzumab, and FAK inhibitors targeting integrins; LDN193189, DMH1, and ISLR modulators targeting BMPs; and PTH (7-33)-CBD targeting PTHrP have shown promising results antagonizing these molecules, but no effect on preventing and managing metastatic fractures has been assessed yet. CONCLUSIONS This paper underscores the importance of advanced molecular biology and transcriptomics in identifying novel therapeutic targets. The integration of these biomarkers with clinical and radiological assessments using artificial intelligence tools could revolutionize the diagnostics and treatment strategies for patients with bone metastases.
Collapse
Affiliation(s)
- Alessandro Bruschi
- Orthopedic and Traumatology UnitIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
- Department of Biomedical and Neuromotor SciencesUniversity of BolognaBolognaItaly
| | - Andrea Sambri
- Orthopedic and Traumatology UnitIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | - Michele Fiore
- Orthopedic and Traumatology UnitIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
- Department of Medical and Surgical SciencesAlma Mater Studiorum University of BolognaBolognaItaly
| | - Elisa Bubbico
- Orthopedic and Traumatology UnitIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | - Cristina Scollo
- Orthopedic and Traumatology UnitIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | - Andrea Pace
- Orthopedic and Traumatology UnitIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | - Renato Zunarelli
- Orthopedic and Traumatology UnitIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | - Andrea Montanari
- Orthopedic and Traumatology UnitIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | - Alberta Cappelli
- Department of RadiologyIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | - Lorenzo Di Prinzio
- Orthopedic and Traumatology UnitIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | - Massimiliano De Paolis
- Orthopedic and Traumatology UnitIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| |
Collapse
|
7
|
Zhang K, Yuan B, Dai X, Chen W, Zhang C, Qiao Y, Cao W, Chen Y, Duan X, Zhang X, Yang W, Li X, Zhao J, Liu K, Dong Z, Lu J. Selection and identification of DNA aptamer binding VDAC1 for tumor tissue imaging and targeted drug delivery. Int J Biol Macromol 2025; 306:141249. [PMID: 39984095 DOI: 10.1016/j.ijbiomac.2025.141249] [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: 12/25/2024] [Revised: 02/14/2025] [Accepted: 02/16/2025] [Indexed: 02/23/2025]
Abstract
Hepatocellular carcinoma (HCC) represents a significant health concern. Identifying novel molecular targets is crucial for clinical diagnosis and targeted treatment of HCC. Aptamers are capable of binding specifically to cancer cells via target protein molecules. Consequently, aptamers are frequently employed to identify novel cancer biomarkers. The invasiveness of tumor cells is closely associated with the recurrence and metastasis of tumors. In this study, the highly invasive Huh7-P3 cells were initially constructed, and subsequently, several aptamers that could specifically recognize Huh7-P3 were developed using cell-based Systematic Evolution of Ligands by Exponential Enrichment (SELEX). The selected aptamer, designated S2-2, demonstrated the capacity to bind to multiple cancer cells. Furthermore, tissue imaging demonstrated that S2-2 exhibited a specific recognition of HCC tissue, while demonstrating no binding to normal tissue. Subsequently, voltage-dependent anion channel 1 (VDAC1) was identified as a potential target for S2-2. Furthermore, Doxorubicin (Dox)-loaded S2-2 was shown to specifically kill target Huh7-P3 cells. In vivo fluorescence imaging revealed that S2-2 was capable of specifically targeting tumors. Importantly, S2-2-Dox enhanced the anti-tumor efficacy of Dox in cell-line-derived xenograft (CDX) model. This study may provide a promising biomarker and molecular target for the clinical diagnosis and targeted therapy of cancers with high VDAC1 expression.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Baoyin Yuan
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Xiaoshuo Dai
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Wei Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Chengjuan Zhang
- Department of Pathology, Henan Cancer Hospital, Zhengzhou University, Zhengzhou, Henan Province 450003, PR China
| | - Yan Qiao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; State Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China
| | - Wenbo Cao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; State Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China
| | - Yihuan Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Xiaoxuan Duan
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Xiaoyan Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; State Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China
| | - Wanjing Yang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; State Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China
| | - Xiang Li
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; State Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China
| | - Jimin Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; State Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; State Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China
| | - Ziming Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; State Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China
| | - Jing Lu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; State Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China.
| |
Collapse
|
8
|
Tan W, Zhu Y, Chen S. Innovative approach to the detection of circulating tumor biomarkers: multi-dimensional application of liposome technology. Lipids Health Dis 2025; 24:160. [PMID: 40295973 PMCID: PMC12036244 DOI: 10.1186/s12944-025-02578-7] [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: 01/28/2025] [Accepted: 04/19/2025] [Indexed: 04/30/2025] Open
Abstract
Malignant tumors represent a significant worldwide health challenge, with elevated morbidity and mortality rates necessitating enhanced early identification and individualized treatment. Liposomes, as biomimetic lipid-based nanovesicles, have developed as a multifaceted platform for detecting and treating malignant tumors due to their excellent biocompatibility, stability, and membrane fusion properties. Circulating tumor markers, such as circulating tumor cells (CTCs), extracellular vesicles (EVs), circulating tumor proteins (CTPs), and circulating tumor nucleic acids (ctNAs), play a key role in early cancer diagnosis, disease progression monitoring, and personalized therapy. Liposome-based platforms enable effective molecular recognition, targeted detection, and signal amplification by targeting circulating tumor biomarkers, significantly increasing the potential for early tumor diagnosis and treatment. This review systematically summarizes advancements in the study of liposomes concerning circulating tumor markers, including applications in targeted recognition, early detection, and disease diagnosis, while discussing present problems and prospective applications of existing technology.
Collapse
Affiliation(s)
- Weichu Tan
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Provincial Key Laboratory of Single-Cell and Extracellular Vesicles, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Yitong Zhu
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Provincial Key Laboratory of Single-Cell and Extracellular Vesicles, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Siting Chen
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Provincial Key Laboratory of Single-Cell and Extracellular Vesicles, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China.
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, 511518, People's Republic of China.
| |
Collapse
|
9
|
Liu Z, Li T, Gu C, Chen C, Tang Z, Feng Y, Zhou C, Xu J, Chen J. Novel agents derived from natural product β-elemene: A second round of design and synthesis to enhance antitumor properties. Bioorg Med Chem 2025; 121:118129. [PMID: 40015120 DOI: 10.1016/j.bmc.2025.118129] [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: 09/20/2024] [Revised: 02/08/2025] [Accepted: 02/20/2025] [Indexed: 03/01/2025]
Abstract
Natural products play a key role in drug discovery and development. The natural sesquiterpene, β-elemene, has been approved as an antitumor drug in China. Despite showing few side effects, the moderate antitumor potency of β-elemene hampers its wide application in clinic. A second round of design and synthesis of β-elemene derivatives was carried out based on our previous prodrug-like ester derivatives. The resulting twenty-nine compounds (except 10c) exhibited enhanced antitumor activity compared with β-elemene and its ester derivative 3. The optimal compound 10a possessed low micromolar antiproliferative activities against three human cancer cell lines (SGC-7901, HeLa, and U87), more potent than positive control cisplatin. The mechanism studies indicate that compound 10a caused arrest of the cell cycle along with inhibition of microtubules, induced apoptosis via a ROS-involved mitochondrial apoptotic pathway, and dampened cell migration and invasion with changes of related protein (MMP-9 and p-FAKY397) expressions. Collectively, the promising antitumor efficacy of compound 10a would make it a potential lead compound in anticancer drug development.
Collapse
Affiliation(s)
- Zhouyan Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Tong Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chenglei Gu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Cheng Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ziwei Tang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yanyan Feng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chen Zhou
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States
| | - Jinyi Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China.
| | - Jichao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
10
|
Matić M, Obradović A, Paunović M, Ognjanović B, Mihailović V, Srećković N, Stanković M. Green-Synthesized Silver Nanoparticles Using Filipendula ulmaria (L.) Maxim. and Salvia verticillata L. Extracts Inhibit Migration and Modulate Redox Homeostasis in Human Breast Cancer Cells via Nrf-2 Signaling Pathway. Antioxidants (Basel) 2025; 14:469. [PMID: 40298802 PMCID: PMC12024124 DOI: 10.3390/antiox14040469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 03/31/2025] [Accepted: 04/08/2025] [Indexed: 04/30/2025] Open
Abstract
Breast cancer is a leading cancer diagnosis for women around the world, with a variable degree of curability. Conventional chemotherapeutic treatments often induce toxicity and damage to healthy tissues, as well as the development of drug resistance, which is why an increasing number of new therapeutic regimens focus on the use of natural products and various modifications of their delivery to target tissues. Silver nanoparticles possess unique physicochemical characteristics, notably their increased surface area, suggesting that they hold significant potential for biomedical applications. This research evaluates the capacity of silver nanoparticles green synthesized with aqueous extracts of Filipendula ulmaria (FUAgNPs) and Salvia verticillata (SVAgNPs) to alter migration and redox homeostasis in the human breast cancer cell line MDA-MB-231. To determine the values of redox homeostasis parameters, the cells were treated with five different concentrations (5, 10, 20, 50, and 100 μg/mL) for 24 h and 72 h, while to test the migratory potential and concentrations of matrix metalloproteinase-9 (MMP-9) and nuclear factor erythroid 2-related factor 2 (Nrf-2), the cells were treated at two concentrations (5 and 50 µg/mL) for 72 h. The obtained results indicate increased production of superoxide anion radicals, malondialdehyde (MDA), and nitrites after the investigated treatment on MDA-MB-231 cells. The treatments induced only a slight elevation in Nrf-2 levels, which correlates with weak de novo synthesis of reduced glutathione (GSH), suggesting that the tested nanoparticles weaken the inherent antioxidative systems of the tested cells. The migration potential of cells was significantly reduced, and MMP-9 concentration was significantly inhibited. Based on the demonstrated antitumor effect, confirmed by the reduced migratory potential of the examined cells and disrupted redox balance, these nanoparticles have potential for additional investigation with the aim of improving the efficacy of antitumor therapy. Also, FUAgNPs and SVAgNPs possess the capacity to be potentially promising novel chemotherapeutic agents against breast cancer progression and metastasis.
Collapse
Affiliation(s)
- Miloš Matić
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia; (M.M.); (M.P.); (B.O.); (M.S.)
| | - Ana Obradović
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia; (M.M.); (M.P.); (B.O.); (M.S.)
| | - Milica Paunović
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia; (M.M.); (M.P.); (B.O.); (M.S.)
| | - Branka Ognjanović
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia; (M.M.); (M.P.); (B.O.); (M.S.)
| | - Vladimir Mihailović
- Department of Chemistry, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia; (V.M.); (N.S.)
| | - Nikola Srećković
- Department of Chemistry, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia; (V.M.); (N.S.)
| | - Milan Stanković
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia; (M.M.); (M.P.); (B.O.); (M.S.)
| |
Collapse
|
11
|
Veilleux C, Khalifa J, Zgheib A, Konan AS, Gaudreault R, Annabi B. Probing into the chemopreventive properties of synthetic 1,3,6-tri-O-galloyl-α-D-glucose (α-TGG) against glioblastoma and triple-negative breast cancer-derived cell models. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2025; 8:100219. [PMID: 40248812 PMCID: PMC12004382 DOI: 10.1016/j.crphar.2025.100219] [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: 12/10/2024] [Revised: 03/13/2025] [Accepted: 03/31/2025] [Indexed: 04/19/2025] Open
Abstract
Inflammation plays a significant role in cancer progression. Chemopreventive strategies against cellular response to pro-inflammatory cues may therefore contribute to inhibit the acquisition of an invasive phenotype. 1,3,6-Tri-O-Galloyl-β-D-Glucose (β-TGG) is a type of gallotannin naturally found in plants like Paeonia lactiflora and Terminalia chebula. Unfortunately, the overall yields of β-TGG extraction require complex purification protocols from plant sources and are relatively low. Here, a new synthetic α-anomer of TGG (α-TGG) was characterized for anti-inflammatory and anticancer biological properties. In vitro pro-inflammatory and epithelial-to-mesenchymal transition (EMT) cues, triggered by phorbol 12-myristate 13-acetate (PMA), concanavalin A (ConA), tumor necrosis factor (TNF) α, and transforming growth factor (TGF) β, were used to screen α-TGG in two highly aggressive human cancer cell models, namely the U87 glioblastoma and the MDA-MB-231 triple-negative breast cancer (TNBC)-derived cells. α-TGG dose-dependently inhibited ConA-mediated activation of the latent matrix metalloproteinase pro-MMP-2 into its active MMP-2 form as well as the ConA- and PMA-mediated cyclooxygenase (COX)-2 expression, two biomarkers of inflammation, in U87 cells. In MDA-MB-231, α-TGG inhibited PMA- and TNFα-mediated induction of pro-MMP-9, a marker of inflammation and invasive phenotype. Finally, in both cell lines, α-TGG further inhibited TGFβ-induced chemotaxis, as well as TGFβ-induced Smad2 phosphorylation and Snail expression, crucial upstream signaling pathway and downstream biomarkers associated with EMT. Collectively, we confirm that α-TGG retained potent anti-inflammatory and anti-invasive pharmacological properties which support its chemopreventive potential.
Collapse
Affiliation(s)
- Carolane Veilleux
- Laboratoire D’Oncologie Moléculaire, Département de Chimie and CERMO-FC, Université du Québec à Montréal, Montréal, QC, H3C 3P8, Canada
| | - Jihane Khalifa
- Département de Chimie, Université Du Québec à Montréal, Montréal, QC, H3C 3P8, Canada
| | - Alain Zgheib
- Laboratoire D’Oncologie Moléculaire, Département de Chimie and CERMO-FC, Université du Québec à Montréal, Montréal, QC, H3C 3P8, Canada
| | - Angélique Sabaoth Konan
- Laboratoire D’Oncologie Moléculaire, Département de Chimie and CERMO-FC, Université du Québec à Montréal, Montréal, QC, H3C 3P8, Canada
| | - Roger Gaudreault
- Département de Chimie, Université Du Québec à Montréal, Montréal, QC, H3C 3P8, Canada
| | - Borhane Annabi
- Laboratoire D’Oncologie Moléculaire, Département de Chimie and CERMO-FC, Université du Québec à Montréal, Montréal, QC, H3C 3P8, Canada
| |
Collapse
|
12
|
Ivaskiene T, Kaspute G, Ramanavicius A, Prentice U. Molecularly Imprinted Polymer Advanced Hydrogels as Tools for Gastrointestinal Diagnostics. Gels 2025; 11:269. [PMID: 40277704 PMCID: PMC12026608 DOI: 10.3390/gels11040269] [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: 03/10/2025] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/26/2025] Open
Abstract
Gastroenterology faces significant challenges due to the global burden of gastrointestinal (GI) diseases, driven by socio-economic disparities and their wide-ranging impact on health and healthcare systems. Advances in molecularly imprinted polymers (MIPs) offer promising opportunities for developing non-invasive, cost-effective diagnostic tools that enhance the accuracy and accessibility of GI disease detection. This research explores the potential of MIP-based sensors in revolutionizing gastrointestinal diagnostics and improving early detection and disease management. Biomarkers are vital in diagnosing, monitoring, and personalizing disease treatment, particularly in gastroenterology, where advancements like MIPs offer highly selective and non-invasive diagnostic solutions. MIPs mimic natural recognition mechanisms, providing stability and sensitivity even in complex biological environments, making them ideal for early disease detection and real-time monitoring. Their integration with advanced technologies, including conducting polymers, enhances their functionality, enabling rapid, point-of-care diagnostics for gastrointestinal disorders. Despite regulatory approval and scalability challenges, ongoing innovations promise to revolutionize diagnostics and improve patient outcomes through precise approaches.
Collapse
Affiliation(s)
- Tatjana Ivaskiene
- State Research Institute Centre for Innovative Medicine, LT-08410 Vilnius, Lithuania; (T.I.); (G.K.)
| | - Greta Kaspute
- State Research Institute Centre for Innovative Medicine, LT-08410 Vilnius, Lithuania; (T.I.); (G.K.)
- Department of Nanotechnology, State Research Institute Center for Physical Sciences and Technology (FTMC), LT-10257 Vilnius, Lithuania
| | - Arunas Ramanavicius
- Department of Nanotechnology, State Research Institute Center for Physical Sciences and Technology (FTMC), LT-10257 Vilnius, Lithuania
- Department of Physical Chemistry, Faculty of Chemistry and Geosciences, Institute of Chemistry, Vilnius University, LT-03225 Vilnius, Lithuania
| | - Urte Prentice
- State Research Institute Centre for Innovative Medicine, LT-08410 Vilnius, Lithuania; (T.I.); (G.K.)
- Department of Nanotechnology, State Research Institute Center for Physical Sciences and Technology (FTMC), LT-10257 Vilnius, Lithuania
- Department of Physical Chemistry, Faculty of Chemistry and Geosciences, Institute of Chemistry, Vilnius University, LT-03225 Vilnius, Lithuania
| |
Collapse
|
13
|
Konstantaraki M, Berdiaki A, Neagu M, Zurac S, Krasagakis K, Nikitovic D. Understanding Merkel Cell Carcinoma: Pathogenic Signaling, Extracellular Matrix Dynamics, and Novel Treatment Approaches. Cancers (Basel) 2025; 17:1212. [PMID: 40227764 PMCID: PMC11987840 DOI: 10.3390/cancers17071212] [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: 03/01/2025] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025] Open
Abstract
Merkel cell carcinoma (MCC) is a rare but aggressive neuroendocrine skin cancer, driven by either Merkel cell polyomavirus (MCPyV) integration or ultraviolet (UV)-induced mutations. In MCPyV-positive tumors, viral T antigens inactivate tumor suppressors pRb and p53, while virus-negative MCCs harbor UV-induced mutations that activate similar oncogenic pathways. Key signaling cascades, including PI3K/AKT/mTOR and MAPK, support tumor proliferation, survival, and resistance to apoptosis. Histologically, MCC consists of small round blue cells with neuroendocrine features, high mitotic rate, and necrosis. The tumor microenvironment (TME) plays a central role in disease progression and immune escape. It comprises a mix of tumor-associated macrophages, regulatory and cytotoxic T cells, and elevated expression of immune checkpoint molecules such as PD-L1, contributing to an immunosuppressive niche. The extracellular matrix (ECM) within the TME is rich in proteoglycans, collagens, and matrix metalloproteinases (MMPs), facilitating tumor cell adhesion, invasion, and interaction with stromal and immune cells. ECM remodeling and integrin-mediated signaling further promote immune evasion and therapy resistance. Although immune checkpoint inhibitors targeting PD-1/PD-L1 have shown promise in treating MCC, resistance remains a major hurdle. Therapeutic strategies that concurrently target the TME-through inhibition of ECM components, MMPs, or integrin signaling-may enhance immune responses and improve clinical outcomes.
Collapse
Affiliation(s)
- Maria Konstantaraki
- Department of Histology-Embryology, Medical School, University of Crete, 71003 Heraklion, Greece; (M.K.); (A.B.)
- Dermatology Department, University Hospital of Heraklion, 71110 Heraklion, Greece;
| | - Aikaterini Berdiaki
- Department of Histology-Embryology, Medical School, University of Crete, 71003 Heraklion, Greece; (M.K.); (A.B.)
| | - Monica Neagu
- Immunology Laboratory, “Victor Babes” National Institute of Pathology, 99-101 Splaiul Independenței, 050096 Bucharest, Romania;
- Pathology Department, Colentina Clinical Hospital, 19-21 Sos Stefan Cel Mare, 020125 Bucharest, Romania;
| | - Sabina Zurac
- Pathology Department, Colentina Clinical Hospital, 19-21 Sos Stefan Cel Mare, 020125 Bucharest, Romania;
- Faculty of Dentistry, University of Medicine and Pharmacy, 8 Eroilor Sanitari Boulevard, 050474 Bucharest, Romania
| | | | - Dragana Nikitovic
- Department of Histology-Embryology, Medical School, University of Crete, 71003 Heraklion, Greece; (M.K.); (A.B.)
| |
Collapse
|
14
|
Shaaban S, Alabdali AYM, Mousa MHA, Ba-Ghazal H, Al-Faiyz YS, Elghamry I, Althikrallah HA, Khatib AOA, Alaasar M, Al-Karmalawy AA. Innovative Multitarget Organoselenium Hybrids With Apoptotic and Anti-Inflammatory Properties Acting as JAK1/STAT3 Suppressors. Drug Dev Res 2025; 86:e70075. [PMID: 40103327 DOI: 10.1002/ddr.70075] [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: 12/18/2024] [Revised: 02/18/2025] [Accepted: 03/06/2025] [Indexed: 03/20/2025]
Abstract
Herein, we report the design, synthesis, and characterization of novel organoselenium (OSe) hybrids (5-19) via modifications of the lead, N-(4-selaneylphenyl)-2-selaneylacetamide. The OSe-based thiazol 9 showed the highest growth inhibition % (GI%) of 64.72% relative to the positive reference doxorubicin (DOX), with a GI% of 79.5%. Furthermore, the novel OSe derivatives showed low GI% values compared to the normal cell lines employed, demonstrating their selectivity. The OSe tethered N-chloroacetamide 5 and Schiff base 19 showed a cytotoxic effect with an IC50 of (25.07 and 11.61 µM), respectively, against the A549 tumor cell line and IC50 of (34.22 and 20.12 µM), respectively, against the HELA cancer cell line. Enzyme-linked immunosorbent assay to study the JAK1 and the STAT3 inhibitory potentials of OSe compounds 5 and 19 in the A549 cancer cells both showed promising inhibitory activities with IC50 values of 25.07 and 11.61 µM, respectively. Protein expression analysis on the A549 cancer cell line on OSe compounds 5 and 19 showed upregulation of P53, BAX, and Caspases 3, 6, 8, and 9 as apoptotic proteins. However, both candidates expressed downregulation of the antiapoptotic proteins (BCL2, MMP2, and MMP9). Moreover, OSe compounds 5 and 19 described the downregulation of the examined inflammatory proteins: COX2, IL-6, and IL-1β. In addition, OSe compound 19 showed potential cell cycle arrest at the G0, S, and G2-M layers, with an increase in cellular levels. Finally, molecular docking studies of OSe compound 19 showed the most promising inhibitory potential toward the JAK1 and STAT3 target receptors, with binding scores and interactions exceeding that of the cocrystallized inhibitor of JAK1.
Collapse
Affiliation(s)
- Saad Shaaban
- Department of Chemistry, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia
| | | | - Mai H A Mousa
- Pharmaceutical Chemistry Department, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, Egypt
| | - Hussein Ba-Ghazal
- Department of Chemistry, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Yasair S Al-Faiyz
- Department of Chemistry, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Ibrahim Elghamry
- Department of Chemistry, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Hanan A Althikrallah
- Department of Chemistry, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Arwa Omar Al Khatib
- Faculty of Pharmacy, Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman, Jordan
| | - Mohamed Alaasar
- Department of Chemistry, Faculty of Science, Cairo University, Giza, Egypt
- Faculty of Natural, Science II, Institute of Chemistry, Martin-Luther University, Halle Saale, Germany
| | - Ahmed A Al-Karmalawy
- Department of Pharmaceutical Chemistry, College of Pharmacy, The University of Mashreq, Baghdad, Iraq
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Horus University-Egypt, New Damietta, Egypt
| |
Collapse
|
15
|
Xie T, Guo J, Wang W. The Long Noncoding RNA Gall Bladder Cancer-Associated Suppressor of Pyruvate Carboxylase Inhibits the Proliferation, Migration, and Invasion of Colorectal Cancer Cells and Induces Their Apoptosis. Biochem Genet 2025; 63:1719-1733. [PMID: 38609669 DOI: 10.1007/s10528-024-10786-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 03/15/2024] [Indexed: 04/14/2024]
Abstract
This study aimed to determine the role of the long noncoding RNA (lncRNA) gall bladder cancer-associated suppressor of pyruvate carboxylase (SOD2-1) in the progression of colorectal cancer (CRC). A total of 23 pairs of specimens, including CRC tissues and adjacent normal tissues, were collected, and the expression of lncRNA SOD2-1 (lnc-SOD2-1) was measured. lnc-SOD2-1 function was examined using HCT15 and HCT116 cells. A lnc-SOD2-1 overexpression vector was designed and transfected into both cell lines. MTS and colony formation assays were used to determine cell viability. Flow cytometry and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling assays were performed to measure apoptosis. Cell migration and invasion were evaluated using the Transwell assay. Migration and invasion markers were validated using quantitative reverse transcription-polymerase chain reaction and western blot analysis. The results indicated that the expression of lnc-SOD2-1 was downregulated in CRC tissues. lnc-SOD2-1 overexpression evidently decreased cell viability and led to the formation of fewer cell colonies. lnc-SOD2-1 overexpression induced ~ twofold higher apoptosis than the control group. lnc-SOD2-1 overexpression reduced the proportion of migratory and invasive cells to 50% and 75% of the control group, respectively. lnc-SOD2-1 overexpression significantly decreased the expression of matrix metalloproteinase-2 and -9. In conclusion, lnc-SOD2-1 may act as a tumor suppressor that inhibits the proliferation, migration, and invasion of CRC cells and induces their apoptosis.
Collapse
Affiliation(s)
- Tingting Xie
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jianian Guo
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wei Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Guangzhou Medical University, No.250 Changgang East Road, Haizhu District, Guangzhou, 510260, China.
| |
Collapse
|
16
|
Ramadan DR, Osman HA, Madhy SA, Teleb M, Darwish AI, Abu-Serie MM, Haiba NS, Khattab SN, Khalil HH. A tailored 4G s-triazine-based dendrimer vehicle for quercetin endowed with MMP-2/9 inhibition and VEGF downregulation for targeting breast cancer progression and liver metastasis. RSC Adv 2025; 15:10426-10441. [PMID: 40182507 PMCID: PMC11967334 DOI: 10.1039/d5ra01588j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Accepted: 03/24/2025] [Indexed: 04/05/2025] Open
Abstract
Motivated by our recent research progress on the exploitation of s-triazine dendritic platforms as bioactive carriers for well-known anticancer agents and/or targeting ligands, we set out to synthesize new rationally designed dendrimers endowed with MMP-2/9 inhibition potential for halting both breast and liver cancer progression with reduced off-target side effects. New three and four generation s-triazine based dendrimers were developed to incorporate potential ZBGs (Zinc Binding Groups) and carboxyl terminal groups to facilitate direct conjugation of anti-cancer drugs (quercetin) and/or targeting ligands (lactobionic acid) through a biodegradable ester bond. Compared to free quercetin (QUR), MTT assay revealed that all the quercetin-coupled dendrimers displayed better anticancer potential (IC50 = 12.690-29.316, 4.137-29.090 μM) against MCF-7 and HepG-2 cancer cells, respectively within their safe doses (EC100 = 134.35-78.44 μM). Conjugation of lactobionic acid and PEG boosted the anticancer potency against both treated cells, improved apoptosis and down regulated MMP-9 and VEGF gene expression levels in both treated cancer cells. Generally, the more branched G4 dendrimer conjugates exhibited a superior overall anticancer performance compared to their respective G3 analogues, except for their MMP-9 inhibition where G3 conjugate appeared to be more potent and more selective than its G4 analogue.
Collapse
Affiliation(s)
- Doaa R Ramadan
- Chemistry Department, Faculty of Science, Alexandria University Alexandria 21321 Egypt
| | - Heba A Osman
- Department of Physics and Chemistry, Faculty of Education, Alexandria University Alexandria Egypt
| | - Somaya Aly Madhy
- Chemistry Department, Faculty of Science, Alexandria University Alexandria 21321 Egypt
| | - Mohamed Teleb
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University Alexandria 21521 Egypt
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University Alexandria 21521 Egypt
| | - A I Darwish
- Department of Physics and Chemistry, Faculty of Education, Alexandria University Alexandria Egypt
| | - Marwa M Abu-Serie
- Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City) Alexandria Egypt
| | - Nesreen S Haiba
- Department of Physics and Chemistry, Faculty of Education, Alexandria University Alexandria Egypt
| | - Sherine N Khattab
- Chemistry Department, Faculty of Science, Alexandria University Alexandria 21321 Egypt
| | - Hosam H Khalil
- Chemistry Department, Faculty of Science, Alexandria University Alexandria 21321 Egypt
| |
Collapse
|
17
|
Khandan M, Khazeei Tabari MA, Rahimi SM, Hassani M, Bagheri A. The effects of flavonoid baicalein on miRNA expressions in cancer: a systematic review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04078-y. [PMID: 40153015 DOI: 10.1007/s00210-025-04078-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/19/2025] [Indexed: 03/30/2025]
Abstract
Baicalein from Scutellaria baicalensis influences miRNA expression in various cancers, affecting key signaling pathways (PI3K/AKT, Wnt/β-catenin, mTOR) and processes like tumor growth, apoptosis, and metastasis. miRNAs, as small non-coding RNAs, play crucial roles in the cancer pathogenesis-associated gene regulations. This study is aimed at systematically reviewing the effects of baicalein on miRNA expression in various cancers. A comprehensive systematic review was conducted following PRISMA guidelines to investigate the impact of baicalein on miRNA expression in cancer. Databases including PubMed, Scopus, and Web of Science were systematically searched using key search terms. Inclusion criteria encompassed studies reporting changes in miRNA expression following baicalein treatment in cancer cell lines and animal models. Data extraction and risk of bias assessment based on SYRCLE's risk of bias tool were performed to ensure methodological rigor and reliability of the findings. Fifteen studies meeting the inclusion criteria were included in the systematic review. Baicalein impacts miRNA expression in cancers like hepatocellular carcinoma, breast, cervical, ovarian, and gastric cancers, suggesting its potential as a multi-cancer therapeutic. Baicalein regulates tumor-related genes (HDAC10, MDM2, Bcl-2/Bax, and Cyclin E1) and signaling molecules (AKT, FOXO3α), affecting cell viability, apoptosis, and cell cycle, indicating targeted therapeutic potential. In vitro and in vivo studies show baicalein inhibits tumor growth, enhances apoptosis, and regulates cell proliferation, supporting its anticancer effects. Baicalein exhibits potential in modulating miRNA expression in cancer, offering avenues for therapeutic intervention. However, methodological rigor in future studies is essential to enhance the reliability and validity of findings. Comprehensive understanding of baicalein's effects on miRNA expression holds promise for developing novel cancer treatment strategies.
Collapse
Affiliation(s)
| | | | | | - Mahmoud Hassani
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abouzar Bagheri
- Imam Khomeini Hospital, Mazandaran University of Medical Sciences, Sari, Iran.
- Immunogenetics Research Center, Department of Clinical Biochemistry and Medical Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
18
|
Gao H, Sun F, Zhang X, Qiao X, Guo Y. The role and application of Coronin family in human tumorigenesis and immunomodulation. Biochim Biophys Acta Rev Cancer 2025; 1880:189304. [PMID: 40154644 DOI: 10.1016/j.bbcan.2025.189304] [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: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
The Coronin family, a class of actin-binding proteins involved in the formation and maintenance of cytoskeleton structural stability, is aberrantly expressed in various tumors, including lung, gastric and head and neck cancers. They can regulate tumor cell metabolism and proliferation through RAC-1 and Wnt/β-Catenin signaling pathways and regulate invasion by influencing the PI3K, PAK4, and MT1-MMP signaling pathways and impacting the actin-network dynamics. In recent years, an increasing number of studies have highlighted the crucial roles of the cytoskeleton and immune modulation in the occurrence and development of tumors. The article delves into the Coronin family's pivotal role in tumor immune evasion, highlighting its modulation of neutrophil, T cell, and vesicular transport functions, as well as its interactions with tumorigenesis related organelles such as the endoplasmic reticulum, Golgi apparatus, mitochondria, and lysosomes. It also summarizes the potential therapeutic applications of the Coronin family in oncology. This review provides valuable insights into the mechanisms through which the Coronin family is implicated in the onset and progression of tumors. It also provides more theoretical foundation for tumor immunotherapy and combination drug therapy.
Collapse
Affiliation(s)
- Huimeng Gao
- Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, Liaoning 110002, China
| | - Fuli Sun
- Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, Liaoning 110002, China; Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Diseases, Shenyang, Liaoning 110002, China
| | - Xuanyu Zhang
- Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, Liaoning 110002, China
| | - Xue Qiao
- Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, Liaoning 110002, China; Department of Central Laboratory, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, Liaoning 110002, China.
| | - Yan Guo
- Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, Liaoning 110002, China; Department of Central Laboratory, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, Liaoning 110002, China.
| |
Collapse
|
19
|
Lo Cicero A, Campora S, Lo Buglio G, Cinà P, Lo Pinto M, Scilabra SD, Ghersi G. Enhancing therapeutic efficacy through degradation of endogenous extracellular matrix in primary breast tumor spheroids. FEBS J 2025. [PMID: 40098313 DOI: 10.1111/febs.70069] [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: 07/18/2024] [Revised: 11/08/2024] [Accepted: 03/10/2025] [Indexed: 03/19/2025]
Abstract
Solid tumors have a complex extracellular matrix (ECM) that significantly affects tumor behavior and response to therapy. Understanding the ECM's role is crucial for advancing cancer research and treatment. This study established an in vitro model using primary cells isolated from a rat breast tumor to generate three-dimensional spheroids. Monolayer cells and spheroid cultures exhibited different protein expression patterns, with primary tumor spheroids presenting an increased level of ECM-related proteins and a more complex extracellular environment. Furthermore, spheroids produce endogenous collagen type I matrix, which is the main component of the tumoral ECM. This matrix is arranged predominantly around the 3D structure, mimicking the conditions of solid tumors. Treatments with recombinant collagenases class II (acting on the linear collagen region) and class I (acting on the 3D-helix region) completely degrade collagen within the spheroid structure. Collagenase pretreatment enhances the accessibility of the anticancer drug doxorubicin to penetrate the core of spheroids and sensitize them to doxorubicin-induced cytotoxicity. Our findings highlight the importance of overcoming drug resistance in breast cancer by targeting the ECM and proposing a novel strategy for improving therapeutic outcomes in solid tumors. By employing a three-dimensional spheroid model, with an endogenous ECM, we can offer more relevant insights into tumor biology and treatment responses.
Collapse
Affiliation(s)
- Alessandra Lo Cicero
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Italy
| | - Simona Campora
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Italy
- Department of Biomedical Engineering Bioscience Center of the University of Cincinnati, OH, USA
| | - Gabriele Lo Buglio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Italy
- Department of Pharmacy, University of Copenhagen, Denmark
| | | | - Margot Lo Pinto
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| | - Simone Dario Scilabra
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| | - Giulio Ghersi
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Italy
- Abiel Srl, Palermo, Italy
| |
Collapse
|
20
|
Yin D, Yang L, Chen Y. Circ_0022587 Regulates Tumor Properties of Human Breast Cancer Cells by Targeting miR-335-5p/Phosphoglycerate Kinase 1 Pathway. J Biochem Mol Toxicol 2025; 39:e70205. [PMID: 40096161 DOI: 10.1002/jbt.70205] [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: 04/18/2024] [Revised: 02/05/2025] [Accepted: 03/02/2025] [Indexed: 03/19/2025]
Abstract
Increasing research indicates that circular RNAs (circRNAs) affect the development of breast cancer (BC) through specific molecular mechanisms. However, there is no data regarding the role of circ_0022587 in BC progression. This investigation aims to reveal the mechanism of circ_0022587 in regulating the malignant progression of BC. The study recruited 27 BC patients undergoing a surgical operation in Nantong First People's Hospital, Affiliated Hospital 2 of Nantong University. Quantitative real-time polymerase chain reaction and RNase R degradation assay were used to verify the circular structure of circ_0022587. 3-(4,5-Dimethylthazol-2-yl)-2,5-diphenyltetrazolium bromide, 5-Ethynyl-2'-deoxyuridine, flow cytometry analysis, transwell and tube formation assays were used to detect the viability, proliferation, apoptosis, invasion and tumor angiogenesis of BC cells, respectively. Glycolysis was evaluated by glycolysis metabolism assays. The associations among miR-335-5p, circ_0022587 and phosphoglycerate kinase 1 (PGK1) were identified by dual-luciferase reporter assays and RNA immunoprecipitation. The effects of circ_0022587 knockdown on tumor growth were evaluated by xenograft nude mouse model assays. The positive expression rates of PGK1, nuclear proliferation marker and matrix metalloprotein 9 were analyzed by immunohistochemistry assays. The results showed that circ_0022587 expression was upregulated in BC tumor tissues and BC cells. Downregulation of circ_0022587 inhibited cell viability, proliferation, invasion ability, tube angiogenesis and glycolysis, and promoted cell apoptosis. Overexpression of circ_0022587 relieved the effect of glycolysis inhibitor (2-Deoxy-D-glucose, 2-DG) on glucose consumption, lactate production, and ATP/ADP ratios. In addition, circ_0022587 interacted with miR-335-5p, and miR-335-5p inhibitors attenuated circ_0022587 silencing-induced effects in BC cells. miR-335-5p bound to PGK1, and PGK1 overexpression relieved miR-335-5p mimics-induced effects in BC cells. Further, circ_0022587 knockdown inhibited tumor formation in vivo. The above results demonstrate that circ_0022587 regulates PGK1 expression by absorbing miR-335-5p, thereby affecting BC development, which may provide a new therapeutic strategy for BC. The study's novelty and innovative potential lie in its discovery of a new regulatory mechanism involving circ_0022587 in the miR-335-5p/PGK1 pathway and its potential clinical relevance. These aspects contribute to the expanding knowledge base of breast cancer research and could potentially lead to improved therapeutic strategies in the future.
Collapse
Affiliation(s)
- Dian Yin
- Department of Oncology, Nantong First People's Hospital, Affiliated Hospital 2 of Nantong University, Nantong, China
| | - Li Yang
- Department of Oncology, Nantong First People's Hospital, Affiliated Hospital 2 of Nantong University, Nantong, China
| | - Ying Chen
- Department of Oncology, Nantong First People's Hospital, Affiliated Hospital 2 of Nantong University, Nantong, China
| |
Collapse
|
21
|
Zhu M, Xu M, Bertheloot D, Brom VC, Sieberath A, Salber J, Welle K, Burger C, Wirtz DC, Wang S, Schildberg FA. Arcyriaflavin A Alleviates Osteoporosis by Suppressing RANKL-Induced Osteoclastogenesis. Int J Mol Sci 2025; 26:2141. [PMID: 40076762 PMCID: PMC11899857 DOI: 10.3390/ijms26052141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Osteoclasts (OCs) are important therapeutic targets in the treatment of osteoporosis. The aim of this study was to explore a novel therapeutic approach for osteoporosis using Arcyriaflavin A (ArcyA), a natural compound derived from the marine invertebrate Eudistoma sp. We systematically evaluated the effects of ArcyA on OC differentiation and function in mouse models using molecular biology assays, cellular function analyses and in vivo animal experiments. We also evaluated the efficacy of ArcyA in human cells. The TRAP staining results provide the first clear evidence of the drug's inhibitory effect, whereby the administration of ArcyA led to a significant reduction in TRAP-positive cells compared to the control group at concentrations that were non-toxic to bone marrow macrophages. Meanwhile, a significant reduction in the number of multinucleated giant cells with more than ten nuclei was observed. Furthermore, similar TRAP staining results were reproduced in human OCs, suggesting that ArcyA has the same effect on OCs derived from human PBMCs. At the molecular level, ArcyA treatment resulted in the downregulation of genes relevant to OC differentiation (NFATc1, cFos and TNFrsf11α), fusion and survival (DCstamp and ATP6v0d2) and resorption function (CTSK, MMP9, integrin β3 and ACP5). A western blot analysis of the corresponding proteins (NFATc1, cFos, CTSK and integrin β3) further confirmed the PCR results. Furthermore, ArcyA-treated OCs produced significantly fewer resorption pits, indicating suppressed bone resorption activity. Consistent with this, in vivo experiments using an ovariectomy (OVX)-induced osteoporosis mouse model showed that ArcyA treatment significantly alleviated bone loss. Mice in the treatment groups had higher BV/TV values, and this therapeutic effect was enhanced in a dose-dependent manner. In addition, our research also showed that IκB could be a potential target for the inhibitory effect of ArcyA. In conclusion, these findings suggest that ArcyA has significant therapeutic potential for the treatment of osteoporosis by inhibiting osteoclastogenesis and bone resorption. Further studies are warranted to explore its clinical applications.
Collapse
Affiliation(s)
- Mengbo Zhu
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Mingwei Xu
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan 030013, China (S.W.)
| | - Damien Bertheloot
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Victoria C. Brom
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Alexander Sieberath
- Department of Experimental Surgery, Centre for Clinical Research, Ruhr-Universität Bochum, 44780 Bochum, Germany
| | - Jochen Salber
- Department of Experimental Surgery, Centre for Clinical Research, Ruhr-Universität Bochum, 44780 Bochum, Germany
- Department of Surgery, Universitätsklinikum Knappschaftskrankenhaus Bochum GmbH, 44892 Bochum, Germany
| | - Kristian Welle
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Christof Burger
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Dieter C. Wirtz
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Shaowei Wang
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan 030013, China (S.W.)
| | - Frank A. Schildberg
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, 53127 Bonn, Germany
| |
Collapse
|
22
|
Burus A, Ozcan M, Canpinar H, Bozdemir O, Zeybek ND, Bayazit Y. The effect of the combination therapy with chlorophyllin, a glutathione transferase P1-1 inhibitor, and docetaxel on triple-negative breast cancer invasion and metastasis in vivo/in vitro. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03929-y. [PMID: 40014124 DOI: 10.1007/s00210-025-03929-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/13/2025] [Indexed: 02/28/2025]
Abstract
The expression of glutathione S-transferase P1 (GSTP1) enzyme increases in cancer cells, leading to anticancer drug resistance. The antioxidant chlorophyllin has an inhibitory effect on GSTP1. In this study, we investigated the effect of chlorophyllin and its combined administration with the chemotherapeutic agent docetaxel on metastatic processes. For this purpose, both the 4T1 triple-negative breast cancer cell line and metastatic animal model were used. The MTT, flow cytometry, and wound healing assays were used to investigate cell viability, cell cycle, and cell migration, respectively. Total gelatinase activity, GST activity, and glutathione levels in cell and liver tissue lysates measured by colorimetric methods. Micrometastases were evaluated histochemically in liver tissue sections. As a result, the coadministration of chlorophyllin and docetaxel significantly inhibited cell migration in vitro. There was a significant decrease in the total gelatinase activity in vivo. We found that only combined treatment reduced the micrometastatic lesions in the liver tissues, though this reduction was not statistically significant. In conclusion, the coadministration of chlorophyllin and docetaxel may have a potential role in controlling metastatic processes by suppressing cell migration, gelatinase activity, and micrometastasis formation in triple-negative breast cancers.
Collapse
Affiliation(s)
- Ayse Burus
- Department of Medical Biochemistry, Hacettepe University Faculty of Medicine, Ankara, Turkey.
| | - Mehmet Ozcan
- Department of Medical Biochemistry, Zonguldak Bulent Ecevit University Faculty of Medicine, Zonguldak, Turkey
| | - Hande Canpinar
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Ozlem Bozdemir
- Department of Stem Cell Sciences, Hacettepe University Graduate School of Health Sciences, Ankara, Turkey
| | - Naciye Dilara Zeybek
- Department of Histology and Embryology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Yasemin Bayazit
- Department of Medical Biochemistry, Hacettepe University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
23
|
Lv Y, Wang L, Zhang Y, Wei D, Hu Y. circDENND4C serves as a sponge for miR-200b to drive non-small cell lung cancer advancement by regulating MMP-9 expression. Front Oncol 2025; 15:1441384. [PMID: 40034591 PMCID: PMC11872906 DOI: 10.3389/fonc.2025.1441384] [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: 05/31/2024] [Accepted: 01/24/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction Lung cancer has a higher incidence and mortality rate than other cancers, especially non-small cell lung cancer (NSCLC), accounting for 85% of the cases. The role of the circDENND4C/miR-200b/matrix metalloproteinase-9 (MMP-9) regulatory axis in NSCLC remains largely unknown. Methods NSCLC cell lines were used to examine the expression of circDENND4C, miR-200b, and MMP-9 via qRT-PCR or Western blot. The target relationship of circDENND4C, miR-200b, and MMP-9 was examined by RNA fluorescence in situ hybridization (RNA-FISH), immunofluorescence (IF), dual-luciferase reporter system, quantitative real-time polymerase chain reaction (qRT-PCR), and Western blot. Then, a cell count kit-8 (CCK-8) experiment, flow cytometry, and migration/invasion assays were performed to assess the biological function of circDENND4C, miR-200b, and MMP-9 by transfecting with their overexpression or knockout plasmids in A549 cells. Finally, the proteins related to cell adhesion and tight junction were further tested by Western blot and IF. Results circDENND4C and MMP-9 were found to be highly expressed in NSCLC cell lines, while miR-200b was lowly expressed in NSCLC cell lines. Moreover, circDENND4C could sponge miR-200b to target MMP-9. Subsequently, it was observed that knockdown of circDENND4C and MMP-9 or the upregulation of miR-200b repressed cell proliferation and cell cycle progression, increased cell apoptosis, and hindered cell migration and invasion. Finally, it was also found that the circDENND4C/miR-200b/MMP-9 regulatory axis might be involved with cell adhesion and tight junction to influence tumor metastasis. Conclusions Altogether, our study reveals a novel regulatory loop in which the circDENND4C/miR-200b/MMP-9 axis may modulate NSCLC progression, indicating potential biomarkers for the diagnosis or treatment of NSCLC.
Collapse
Affiliation(s)
- Yaming Lv
- Department of Respiratory Medicine, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- Department of Respiratory Medicine, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Lan Wang
- Department of Respiratory Medicine, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- Department of Respiratory Medicine, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Yunhui Zhang
- Department of Respiratory Medicine, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- Department of Respiratory Medicine, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Dong Wei
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yajie Hu
- Department of Respiratory Medicine, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- Department of Respiratory Medicine, The First People’s Hospital of Yunnan Province, Kunming, China
| |
Collapse
|
24
|
Wang A, Yan S, Jiang W, Chen X, Huang Y, Zu X, Du X, Fan L, Liu J, Sun G. Endoplasmic reticulum stress-related CLIP4 plays a procarcinogenic role in hepatocellular carcinoma: an integrated analysis. BMC Cancer 2025; 25:211. [PMID: 39920578 PMCID: PMC11804071 DOI: 10.1186/s12885-025-13537-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 01/15/2025] [Indexed: 02/09/2025] Open
Abstract
OBJECTIVE To explore the potential of endoplasmic reticulum stress (ERS)-associated protein CLIP4 as a biomarker for hepatocellular carcinoma (HCC) and the underlying mechanism. METHODS TCGA public database and a tissue microarray were used to investigate the molecular characteristics of CLIP4 and its association with disease. TCGA-LIHC dataset was used for single-gene differential expression analysis, single-gene correlation analysis, functional enrichment analysis, immune infiltration analysis, and DNA methylation analysis. RNA-seq, immunohistochemistry, western blotting, and RT-qPCR were used to verify the effect of ERS on CLIP4 expression. Public databases and miRNA-seq data were used to explore the TF-miRNA-CLIP4 regulatory network. CCK-8, colony formation, EdU staining, wound-healing, Transwell, western blotting and RT-qPCR were used to detect the effects of CLIP4 on the proliferation, migration and epithelial-mesenchymal transition (EMT) of HCC cells. RESULTS Analysis of TCGA datasets and tissue microarrays demonstrated that elevated CLIP4 expression was associated with poor prognosis in HCC. Enrichment analysis revealed that CLIP4 is involved in the immune response, cell adhesion, and EMT. There was a positive correlation between CLIP4 expression and the infiltration of the majority of immune cells, immunomodulators, and chemokines. Furthermore, the DNA methylation pattern of CLIP4 was found to have significant prognostic value. ERS was found to significantly upregulate CLIP4 expression. In addition, the ERS-RELA-miR-222-5p-CLIP4 transcriptional network was constructed to clarify the role of CLIP4. Cell function experiments confirmed that it promotes the proliferation, migration, and EMT of HCC cells. CONCLUSIONS CLIP4 is a potential immune-related oncogenic molecule in HCC. ERS regulates the expression of CLIP4, and CLIP4 promotes the proliferation, migration, and EMT of HCC cells.
Collapse
Affiliation(s)
- Anqi Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, China
| | - Sitong Yan
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, China
- Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Weijia Jiang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, China
| | - Xiang Chen
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, China
| | - Yuhan Huang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, China
| | - Xiangyu Zu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, China
| | - Xiao Du
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, China
| | - Lulu Fan
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, China
| | - Jiatao Liu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Guoping Sun
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, China.
| |
Collapse
|
25
|
Jadhao M, Hsu SK, Deshmukh D, Liu PF, Weng SF, Chen YF, Li CY, Wang CY, Tsai EM, Wang LF, Chiu CC. Prolonged DEHP exposure enhances the stemness and metastatic potential of TNBC cells in an MSI2-dependent manner. Int J Biol Sci 2025; 21:1705-1729. [PMID: 39990676 PMCID: PMC11844279 DOI: 10.7150/ijbs.101598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 01/02/2025] [Indexed: 02/25/2025] Open
Abstract
Di(2-ethylhexyl) phthalate (DEHP) is a commonly used plasticizer, and human exposure to phthalates is a major health concern. DEHP, which is widely recognized as an endocrine disruptor, is associated with an increased risk of several diseases, including breast cancer. Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer, and metastasis is the leading cause of TNBC-related mortality. However, the correlation between DEHP exposure and TNBC metastasis remains elusive. In the present study, we found that prolonged DEHP treatment enhanced the migration and invasion of TNBC cells both in vitro and in vivo. Mechanistically, DEHP exposure induced Musashi RNA binding protein 2 (MSI2) overexpression, which subsequently activated the PI3K/Akt/NF-κB/MMP-9 axis to augment metastatic potential. MSI2 also promoted stemness. Interestingly, we identified a novel function of MSI2 in regulating the expression, distribution, and polarization of vimentin that is independent of its conventional RNA binding and translation regulation. Genetic knockdown of MSI2 potently abolished DEHP-mediated TNBC progression. Moreover, MSI2 depletion inhibited lung metastasis in metastatic mouse models but did not affect proliferation or tumor size. Intriguingly, miR-155-5p downregulation was observed after DEHP exposure, while mimic miR-155-5p treatment inhibited DEHP-induced TNBC migration, accompanied by reduced expression of MSI2 and vimentin. These findings suggested an inverse relationship between miR-155-5p levels and MSI2 expression. Taken together, MSI2 might serve as a potential therapeutic target and function as a prognostic biomarker for TNBC patients.
Collapse
Affiliation(s)
- Mahendra Jadhao
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati 45220, OH, USA
| | - Sheng-Kai Hsu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Dhanashri Deshmukh
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati 45220, OH, USA
| | - Pei-Feng Liu
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Shih-Feng Weng
- Department of Healthcare Administration and Medical Informatics, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yih-Fung Chen
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chia-Yih Wang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Eing-Mei Tsai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Li-Fang Wang
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
- Center for Cancer Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| |
Collapse
|
26
|
Tran NM, Truong AT, Nguyen DT, Dang TT. Profiling Pro-Inflammatory Proteases as Biomolecular Signatures of Material-Induced Subcutaneous Host Response in Immuno-Competent Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2309709. [PMID: 39630111 PMCID: PMC11792001 DOI: 10.1002/advs.202309709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 09/19/2024] [Indexed: 02/05/2025]
Abstract
Proteases are important modulators of inflammation, but they remain understudied in material-induced immune response, which is critical to clinical success of biomedical implants. Herein, molecular expression and proteolytic activity of three distinct proteases, namely neutrophil elastase, matrix metalloproteinases, cysteine cathepsins (cathepsin-K and cathepsin-B) are comprehensively profiled, in the subcutaneous host response of immuno-competent mice against different biomaterial implants. Quantitative non-invasive monitoring with activatable fluorescent probes reveals that different microparticulate materials induce distinct levels of protease activity with degradable poly(lactic-co-glycolic) acid inducing the strongest signal compared to nondegradable materials such as polystyrene and silica oxide. Furthermore, protein expression of selected proteases, attributable to both their inactive and active forms, notably deviates from their activities associated only with their active forms. Protease activity exhibits positive correlations with protein expression of pro-inflammatory cytokines tumor necrosis factor α and interleukin 6 but negative correlation with pro-fibrotic cytokine transforming growth factor β1. This study also demonstrates the predictive utility of protease activity as a non-invasive, pro-inflammatory parameter for evaluation of the anti-inflammatory effects of model bioactive compounds on material-induced host response. Overall, the findings provide new insights into protease presence in material-induced immune responses, facilitating future biomaterial assessment to evoke appropriate host responses for implant applications.
Collapse
Affiliation(s)
- Nam M.P. Tran
- School of ChemistryChemical Engineering and BiotechnologyNanyang Technological University70 Nanyang DriveSingapore637459Singapore
| | - Anh T.H. Truong
- School of ChemistryChemical Engineering and BiotechnologyNanyang Technological University70 Nanyang DriveSingapore637459Singapore
| | - Dang T. Nguyen
- School of ChemistryChemical Engineering and BiotechnologyNanyang Technological University70 Nanyang DriveSingapore637459Singapore
| | - Tram T. Dang
- School of ChemistryChemical Engineering and BiotechnologyNanyang Technological University70 Nanyang DriveSingapore637459Singapore
| |
Collapse
|
27
|
Bie L, Chen G, Lei X, Xiao F, Xu Z, Xiang Z, Lu Z, Jiang X. B4GALNT1 Regulates Hepatocellular Carcinoma Cell Proliferation and Apoptosis via the PI3K-AKT-mTOR Pathway. J Clin Lab Anal 2025; 39:e25155. [PMID: 39829207 PMCID: PMC11848214 DOI: 10.1002/jcla.25155] [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: 09/01/2024] [Revised: 12/15/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a ubiquitous malignancy linked to significant mortality. The abnormal expression of β-1,4-N-acetyl-galactosaminyltransferase 1 (B4GALNT1) seemed to be implicated in tumorigenesis. Nonetheless, this enzyme's roles in HCC are unclear. METHODS By analyzing the TCGA_LIHC, GSE77509, and GSE135631 datasets, the levels of B4GALNT1 expression in HCC and surrounding non-cancerous tissues were compared. The prognostic implications of B4GALNT1 were assessed using the Cox regression analysis (CRA). The relationship of B4GALNT1 mutations with CpG island methylation levels and prognosis was examined by analyzing the cBioPortal and MethSurv databases. We sifted the evidence of B4GALNT1 expression correlating with 28 immune cell types' infiltration by harnessing the "GSVA" R package. To delve into the influences of genes associated with B4GALNT1 on HCC, we implemented gene set enrichment analysis (GSEA). We constructed a lentiviral vector expressing B4GALNT1 and knocked down B4GALNT1 in HepG2 cells. The resulting effects on HCC cell proliferation and apoptosis were analyzed via cell proliferation assays and flow cytometry. RESULTS HCC tissues presented significant B4GALNT1 overexpression relative to surrounding non-cancerous tissues, marking it as a standalone risk factor for HCC progression. Methylation levels of two CpG islands were high, suggesting poor prognosis. It was detectable that B4GALNT1 expression interrelated with the infiltration extent of natural killer T cells in HCC tissues. B4GALNT1-fueled cell proliferation and enhanced resistance to apoptosis in HCC cells. CONCLUSION B4GALNT1 is a strong regulator of HCC progression and holds promise as a marker for prognosis and a hallmark for therapy in HCC.
Collapse
Affiliation(s)
- Lihan Bie
- Department of Laboratory MedicineThe Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Guangquan Chen
- Shanghai Key Laboratory of Maternal‐Fetal Medicine, Department of Fetal Medicine and Prenatal Diagnosis Center, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Xin Lei
- Department of Laboratory MedicineThe Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Feng Xiao
- Department of PathologyThe Seventh People's Hospital Affiliated to the Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Zheng Xu
- Department of Laboratory MedicineThe Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Zhouhong Xiang
- Department of Laboratory MedicineThe Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Zhicheng Lu
- Department of Laboratory MedicineThe Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Xiudi Jiang
- Department of Laboratory MedicineThe Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
28
|
Chen Z, Yao Y, Gao J. LncRNA ZEB1-AS1 promotes the proliferation and migration of non-small cell lung cancer by activating epithelial-mesenchymal transition with STAT3. Transl Cancer Res 2025; 14:584-594. [PMID: 39974382 PMCID: PMC11833408 DOI: 10.21037/tcr-2024-2276] [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: 11/15/2024] [Accepted: 01/08/2025] [Indexed: 02/21/2025]
Abstract
Background Lung cancer is the most common cause of cancer-related death worldwide. Non-small cell lung cancer (NSCLC) is the main type of lung cancer. Long non-coding RNA ZEB1 antisense 1 (lncRNA ZEB1-AS1) is derived from the promoter region of the transcriptional repressor ZEB1. In bladder cancer and glioblastoma, lncRNA ZEB1-AS1 promotes the expression of ZEB1 and cancer progression, and is associated with a poor prognosis. However, its role in NSCLC tumor progression remains unclear. This study aims to investigate its possible role in NSCLC tumor progression. Methods In this study, overexpressed and silenced lncRNA NSCLC cell lines of ZEB1-AS1 were constructed, epithelial-mesenchymal transition (EMT)-related proteins were detected, and the invasion and migration abilities of the cells were examined. Moreover, the radioimmunoprecipitation (RIP) assay was used to examine whether the increase in the STAT3 protein level caused by ZEB1-AS1 overexpression was based on the promotion of STAT3 messenger RNA (mRNA) translation by AUF1, and the dual-luciferase assay was used to verify the results. Results The overexpression of ZEB1-AS1 increased the protein levels of ZEB1 and STAT3, promoted the occurrence of EMT, and enhanced the invasion and migration abilities of lung cancer cells. The RIP results showed that both lncRNA ZEB1-AS1 and ZEB1 mRNA bind to AUF1, but no binding between AUF1 and STAT3 mRNA was detected. The bioinformatics analysis and the results of the dual-luciferase experiments showed that STAT3 was the target gene of microRNA 519d (miRNA519d), and that lncRNA ZEB1-AS1 also binds to miRNA519d. Conclusions LncRNA ZEB1-AS1 formed the competing endogenous RNA (ceRNA) regulatory network of lncRNA ZEB1-AS1~miRNA519d~STAT3 as the molecular sponge, and promoted the expression of STAT3, thus promoting the occurrence of EMT in lung cancer cells.
Collapse
Affiliation(s)
- Zhengjin Chen
- Department of Respiratory, Nanhu District Central Hospital of Jiaxing, Jiaxing, China
| | - Yangwei Yao
- Pneumology Department, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, China
| | - Jingpeng Gao
- Pneumology Department, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, China
| |
Collapse
|
29
|
Yang Y, Sun H, Yu H, Wang L, Gao C, Mei H, Jiang X, Ji M. Tumor-associated-fibrosis and active collagen-CD44 axis characterize a poor-prognosis subtype of gastric cancer and contribute to tumor immunosuppression. J Transl Med 2025; 23:123. [PMID: 39871345 PMCID: PMC11773867 DOI: 10.1186/s12967-025-06070-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 01/04/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Tumor-associated fibrosis modifies the tumor microenvironment (TME), hinders the infiltration and activity of cytotoxic immune cells, and is a critical pathological process leading to the ineffectiveness of tumor immunotherapy in gastric cancer (GC). However, the specific mechanisms and interventions are yet to be fully explored. METHODS Our study included 375 gastric cancer samples from TCGA, 1 single-cell RNA sequencing (scRNA-seq) dataset comprising of 15 gastric cancer samples from GEO, 19 cohorts of immunotherapy and 2 GWAS datasets. Consensus clustering identified a gastric cancer subtype characterized primarily by fibrosis, and various methods such as pseudotime analysis, CellChat analysis and Colocalization analysis were used to explore its mechanisms. RESULTS A subtype of gastric cancer was identified with poor prognosis, characterized by higher malignancy, drug resistance, and poor immune infiltration, associated with elevated expression of genes related with Extracellular matrix (ECM). Single-cell transcriptome analysis showed active Collagen-CD44 signaling axis between cancer-associated fibroblasts (CAFs) and immune cells in gastric cancer, with ECM-related genes upregulated during tumor progression. The expression of CD44 was significantly elevated in the subtype, associated with poor prognosis and tumor immune suppression in gastric cancer, potentially involved in the recruitment of immunosuppressive cells such as M2 macrophages and regulatory T cells (Tregs) and the upregulation of multiple immune checkpoints including PD-1/PD-L1. CONCLUSION Our study identified a new subtype of gastric cancer, revealing that fibrosis is a critical mechanism driving immune suppression in gastric cancer and emphasizing the central role of the Collagen-CD44 signaling axis. The Collagen-CD44 signaling axis has the potential to serve as a novel therapeutic target for gastric cancer by enhancing immune cell-mediated tumor suppression. By combining it with immune checkpoint inhibitors (ICIs), it may improve the efficacy of immunotherapy for gastric cancer and offer new hope for treatment.
Collapse
Affiliation(s)
- Yingqi Yang
- The Second School of Clinical Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Haohan Sun
- The Second School of Clinical Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Hongkai Yu
- The Second School of Clinical Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Luyao Wang
- The Second School of Clinical Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Chang Gao
- The Second School of Clinical Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Haokun Mei
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Xiaomeng Jiang
- Department of Gastroenterology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211100, China.
| | - Minghui Ji
- School of Nursing, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
30
|
Shetty SS, Ram Padam KS, Sharma M, Kudva A, Patel P, Radhakrishnan R. Novel transcripts of EMT driving the malignant transformation of oral submucous fibrosis. Sci Rep 2025; 15:3294. [PMID: 39865173 PMCID: PMC11770107 DOI: 10.1038/s41598-025-87790-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 01/22/2025] [Indexed: 01/28/2025] Open
Abstract
Oral submucous fibrosis (OSF) is a chronic, progressive, and fibrotic condition of the oral mucosa that carries an elevated risk of malignant transformation. We aimed to identify and validate novel genes associated with the regulation of epithelial-to-mesenchymal transition (EMT) in OSF. Genes regulating EMT were identified through differential gene expression analysis, using a LogFC threshold of -1 and + 1 and a padj value < 0.05, based on data from GEO datasets and the TCGA-HNSC datasets. The curated EMT genes were correlated with functional cancer states and subjected to clustering to identify candidate genes. Integration of bioinformatics and proteomics led to the discovery of the EMT genes MMP9, SPARC, and ITGA5 as novel candidates. Comprehensive pathway and immunohistochemical analyses confirmed their roles in regulating EMT in OSF, oral squamous cell carcinoma (OSCC), and OSF-associated squamous cell carcinoma (OSFSCC). The significant roles of MMP9, SPARC, and ITGA5 in fibrosis and malignancy suggest a novel mechanism in which fibrosis-associated type 2 EMT undergoes transition to type 3 EMT, driving OSF towards malignancy.
Collapse
Affiliation(s)
- Smitha Sammith Shetty
- Department of Oral and Maxillofacial Pathology and Microbiology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Kanaka Sai Ram Padam
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Mohit Sharma
- Department of Oral Pathology, SGT Dental College Hospital & Research Institute, Gurugram, 122505, Haryana, India
| | - Adarsh Kudva
- Department of Oral and Maxillofacial Surgery, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Pratik Patel
- Sangee Oral Pathology Center, Haripura, Surat, 395003, Gujarat, India
| | - Raghu Radhakrishnan
- Department of Oral and Maxillofacial Pathology and Microbiology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, 576104, India.
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, S102TA, UK.
- Unit of Oral and Maxillofacial Pathology, Oman Dental College, P.O Box 835, Muscat, Wattayah 116, Oman.
| |
Collapse
|
31
|
Liu Y, Zhang G, Ma Y, Ma M, Jiang X. Eugenol inhibits NEAT1 as a ceRNA in pre-cancerous breast lesions. Heliyon 2025; 11:e41353. [PMID: 39811361 PMCID: PMC11732537 DOI: 10.1016/j.heliyon.2024.e41353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
Objective Eugenol (EU) from cloves is highly effective against different tumors. The long noncoding ribonucleic acids (lncRNAs), which play a role of competing endogenous RNAs (ceRNAs), suppress microRNAs (miRNAs) involved in post-transcriptional regulatory networks. The present work focused on analyzing how EU affected pre-cancerous breast lesions (PBL). Methods Initially, the gene expression profiles of patients (n = 880) in the National Center for Biotechnology Information (NCBI) database were analyzed. Further, we established a lncRNA-miRNA-mRNA ceRNA network through bioinformatics analysis and investigated mechanistic roles of lncRNAs as ceRNAs and the anti-tumor effect of EU using MCF-10AT cells in vitro as well as PBL model rats in vivo. Besides, Nuclear Paraspeckle Assembly Transcript 1 (NEAT1), miR-383-5p, miR-9-5p, matrix metalloproteinase-9 (MMP-9), and vascular endothelial growth factor-A (VEGF-A) expression was examined through quantitative reverse transcription polymerase chain reaction (RT-qPCR), Western blotting, and immunohistochemical staining analyses. Results There were altogether 1162 mRNAs, 81 miRNAs, and 26 lncRNAs recognized as trend genes in breast cancer (BC) and pre-cancerous BC (pBC), constructing the ceRNA network using 3 lncRNAs, 3 miRNAs, and 38 mRNAs. It was observed that NEAT1, miR-383-5p, miR-9-5p, VEGF-A, and MMP-9 were downregulated in breast tumor cells in accordance with bioinformatics analysis. EU suppressed MCF-10AT cell growth, decreasing the NEAT1, VEGF-A, and MMP-9 levels and increasing miR-383-5p and miR-9-5p expressions in vitro and in vivo. Conclusion In summary, the EU reduced the VEGF-A and MMP-9 expressions via NEAT1-mediated miR-383-5p and miR-9-5p against PBL, indicating that the EU may be a promising external drug to act against PBL.
Collapse
Affiliation(s)
- Yusheng Liu
- Department of Traditional Chinese Medicine, Yangjiang People's Hospital, Yangjiang, Guangdong, 529525, China
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, 510515, China
| | - Guijuan Zhang
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, 510515, China
| | - Yi Ma
- Institute of Biomedicine and Department of Cellular Biology, Jinan University, Guangzhou, Guangdong, 510515, China
| | - Min Ma
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, 510515, China
| | - Xuefeng Jiang
- Department of Traditional Chinese Medicine, Yangjiang People's Hospital, Yangjiang, Guangdong, 529525, China
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, 510515, China
| |
Collapse
|
32
|
Gupta R, Roy D, Ghosh A, Begum Y, Ghosh D, Swarnakar S. Mebendazole Exerts Anticancer Activity in Ovarian Cancer Cell Lines via Novel Girdin-Mediated AKT/IKKα/β/NF-κB Signaling Axis. Cells 2025; 14:113. [PMID: 39851541 PMCID: PMC11763501 DOI: 10.3390/cells14020113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 01/02/2025] [Accepted: 01/09/2025] [Indexed: 01/26/2025] Open
Abstract
Mebendazole (MBZ), a benzimidazole anthelmintic and cytoskeleton-disrupting compound, exhibits antitumor properties; however, its action on ovarian cancer (OC) is not clearly understood. This study evaluates the effect of MBZ on OC cell lines OVCAR3 and OAW42, focusing on cell proliferation, migration, invasion, and cancer stemness. The underlying mechanisms, including cytoskeletal disruption, epithelial-mesenchymal transition (EMT), and signaling pathways, were explored. MBZ inhibited OVCAR3 and OAW42 cell proliferation in a dose- and time-dependent manner. Additionally, MBZ significantly impedes migration, spheroid invasion, colony formation, and stemness. In addition, it reduced actin polymerization and down-regulated CSC markers (e.g., CD24, CD44, EpCAM). Moreover, MBZ suppressed MMP-9 activity and inhibited the EMT marker as judged by decreased N-Cadherin and Vimentin and increased E-Cadherin. Furthermore, MBZ induced G2/M cell cycle arrest by modulating Cyclin B1, CDC25C, and WEE1. Also, it triggered apoptosis by disrupting mitochondrial membrane potential. Mechanistic studies revealed a significant downregulation of Girdin, an Akt modulator, along with reduced p-Akt, p-IKKα/β, and p-NF-κB, indicating MBZ's novel mechanism of action through the Girdin-mediated Akt/IKKα/β/NF-κB signaling axis. Thus, by targeting Girdin, MBZ presents a promising repurposed therapeutic strategy to inhibit cancer cell proliferation and metastasis in ovarian cancer.
Collapse
Affiliation(s)
- Rahul Gupta
- Infectious Diseases & Immunology Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Jadavpur, Kolkata 700032, India or (R.G.); or (Y.B.)
| | - Dipanjan Roy
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, Kolkata 700054, India;
| | - Arijit Ghosh
- Department of Molecular Biology, Netaji Subhash Chandra Bose Cancer Research Institute, Kolkata 700094, India;
| | - Yasmin Begum
- Infectious Diseases & Immunology Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Jadavpur, Kolkata 700032, India or (R.G.); or (Y.B.)
| | - Dipanjan Ghosh
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, Kolkata 700054, India;
| | - Snehasikta Swarnakar
- Infectious Diseases & Immunology Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Jadavpur, Kolkata 700032, India or (R.G.); or (Y.B.)
| |
Collapse
|
33
|
Hao X, Gao Z, Hu M. Anti-tumor role and molecular mechanism of vanillic acid. Discov Oncol 2025; 16:20. [PMID: 39775208 PMCID: PMC11711440 DOI: 10.1007/s12672-025-01746-6] [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: 09/11/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025] Open
Abstract
Vanillic Acid (VA) is an aromatic acid extracted from traditional Chinese medicine such as Angelica sinensis and Panax ginseng, which has demonstrated potent anti-cancer activity, inhibiting the onset and progression of various malignant tumors. This review highlights the principal mechanism by which VA exerts its anticancer activity, including apoptosis induction, specifically promoting the generation of intracellular reactive oxygen species (ROS), which in turn triggers mitochondrial apoptosis. Furthermore, VA disrupts the cancer cell cycle, arresting most cancer cells at the G1 phase, curtails cell migration, invasion, angiogenesis, and potentiates the therapeutic efficacy of chemotherapeutic drugs, all while minimizing adverse reactions. This paper offers a comprehensive review of VA's anti-tumor effects and underlying mechanisms, aiming to provide some references for scientists and clinical physicians in the research of anti-tumor therapeutic strategies.
Collapse
Affiliation(s)
- Xunxing Hao
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shangdong, China
| | - Zhixiao Gao
- Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Mingzhe Hu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
34
|
Liu B, Tian H, Momeni MR. The interplay of exercise and green tea: a new road in cancer therapy. Cancer Cell Int 2025; 25:6. [PMID: 39773739 PMCID: PMC11705833 DOI: 10.1186/s12935-024-03632-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025] Open
Abstract
Exercise is one of the most important activities for every individual due to its proven health beneficials. Several investigations have highlighted the advantageous impacts of aerobic exercise, largely attributed to its capacity to enhance the body's capability to defend against threats against oxidative stress. The information currently accessible suggests that adding regular aerobic exercise to a daily routine greatly decreases the chances of developing serious cancer and passing away. An unevenness in the levels of free radicals and the body's antioxidant defenses, made up of enzyme and non-enzyme antioxidants, results in oxidative pressure. Generally, an imbalance in the levels of oxidative stress triggers the creation of harmful reactive oxygen or nitrogen compounds, causing the development or progression of numerous ailments, including cancer. The equilibrium between pro-oxidant and antioxidant substances is a direct indicator of this imbalance. Green tea and its derivatives are rich sources of bioactive substances such as flavonoids and polyphenols which possess antioxidant abilities. Moreover, modulation of epigenetic targets as well as inflammatory pathways including ERK1/2 and NF-κB are other proposed mechanisms for its antioxidant activity. Recent studies demonstrate the promise of green tea as an antioxidant, showing its ability to decrease the likelihood of developing cancer by impacting actions like cell growth, blood vessel formation, and spread of cancer cells. This summary will concentrate on the complex network of different pathways related to physical activity and consumption of green tea. In particular, the focus of this research will be on examining how oxidative stress contributes to health and investigating the potential antioxidant properties of green tea, and the interconnected relationship between exercise and green tea in the treatment of cancer. Elucidation of these different pathways would help scientists for development of better therapeutic targets and further increase of current anticancer agents efficiency.
Collapse
Affiliation(s)
- Bing Liu
- Henan University of Chinese Medicine, Zhengzhou, 450000, Henan, China
| | - Heyu Tian
- Henan University of Chinese Medicine, Zhengzhou, 450000, Henan, China.
| | | |
Collapse
|
35
|
George J. Zymography: A Simple and Powerful Tool for the Assessment of MMP-2 and MMP-9 in Pathological Conditions. Methods Mol Biol 2025; 2918:187-199. [PMID: 40261623 DOI: 10.1007/978-1-0716-4482-9_15] [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: 04/24/2025]
Abstract
Zymography is an electrophoretic technique to measure the proteolytic activity of native enzymes on a nonreducing sodium dodecyl sulfate polyacrylamide gel. It is a simple and powerful tool to assess the amount of various proteases present in both physiological and pathological conditions. The concerned protease degrades the protein substrate that is incorporated with the gel and resolves during the incubation period. Staining with Coomassie brilliant blue (CBB) reveals the sites of proteolysis as clear white bands. The intensity and area of the bands are linearly related to the amount of protease present in the loaded sample. Matrix metalloproteinase-2 (MMP-2) and matrix metalloproteinase-9 (MMP-9), also known as gelatinases, have the indigenous property to digest gelatin and several other protein molecules present in the extracellular matrix. Here, we describe the detailed protocols and methods of zymography, with a special emphasis on the determination of gelatinases present in conditioned culture media and tissue extracts.
Collapse
Affiliation(s)
- Joseph George
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa, Japan.
| |
Collapse
|
36
|
Yang J, Li Y, Zhang Y, Xu L, Wang J, Xing F, Song X. Unraveling the Core Components and Critical Targets of Houttuynia cordata Thunb. in Treating Non-small Cell Lung Cancer through Network Pharmacology and Multi-omics Analysis. Curr Pharm Des 2025; 31:540-558. [PMID: 39440769 PMCID: PMC12079317 DOI: 10.2174/0113816128330427241017110325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/13/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVE This study aimed to preliminary explore the molecular mechanisms of Houttuynia cordata Thunb. (H. cordata; Saururaceae) in treating non-small cell lung cancer (NSCLC), with the goal of screening drug potential targets for clinical drug development. METHODS This study employed a multi-omics and multi-source data integration approach to identify potential therapeutic targets of H. cordata against NSCLC from the TCMSP database, GEO database, BioGPS database, Metascape database, and others. Meanwhile, target localization was performed, and its possible mechanisms of action were predicted. Furthermore, dynamics simulations and molecular docking were used for verification. Multi-omics analysis was used to confirm the selected key genes' efficacy in treating NSCLC. RESULTS A total of 31 potential therapeutic targets, 8 key genes, and 5 core components of H. cordata against NSCLC were screened out. These potential therapeutic targets played a therapeutic role mainly by regulating lipid and atherosclerosis, the TNF signaling pathway, the IL-17 signaling pathway, and others. Molecular docking indicated a stable combination between MMP9 and quercetin. Finally, through multi-omics analysis, it was found that the expression of some key genes was closely related not only to the progression and prognosis of NSCLC but also to the level of immune infiltration. CONCLUSION Through comprehensive network pharmacology and multi-omics analysis, this study predicts that the core components of H. cordata play a role in treating NSCLC by regulating lipid and atherosclerosis, as well as the TNF signaling pathway. Among them, the anti-NSCLC activity of isoramanone is reported for the first time.
Collapse
Affiliation(s)
- Jinyan Yang
- College of Life Science, Xinyang Normal University, Xinyang 464000, China
| | - Yang Li
- Department of Ultrasound, Xinyang Central Hospital, Xinyang 464000, China
| | - Yan Zhang
- College of Life Science, Xinyang Normal University, Xinyang 464000, China
| | - Ling Xu
- College of Life Science, Xinyang Normal University, Xinyang 464000, China
| | - Jiahui Wang
- College of International Education, Xinyang Normal University, Xinyang 464000, China
| | - Feng Xing
- College of Life Science, Xinyang Normal University, Xinyang 464000, China
| | - Xinqiang Song
- College of Life Science, Xinyang Normal University, Xinyang 464000, China
- Medical College, Xinyang Normal University, Xinyang 464000, China
| |
Collapse
|
37
|
Amini J, Zafarjafarzadeh N, Ghahramanlu S, Mohammadalizadeh O, Mozaffari E, Bibak B, Sanadgol N. Role of Circular RNA MMP9 in Glioblastoma Progression: From Interaction With hnRNPC and hnRNPA1 to Affecting the Expression of BIRC5 by Sequestering miR-149. J Mol Recognit 2025; 38:e3109. [PMID: 39401767 DOI: 10.1002/jmr.3109] [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: 05/01/2024] [Revised: 07/29/2024] [Accepted: 09/27/2024] [Indexed: 01/04/2025]
Abstract
Glioblastoma multiforme (GBM) presents a significant challenge in neuro-oncology due to its aggressive behavior and self-renewal capacity. Circular RNAs (circRNAs), a subset of non-coding RNAs (ncRNAs) generated through mRNA back-splicing, are gaining attention as potential targets for GBM research. In our study, we sought to explore the functional role of circMMP9 (circular form of matrix metalloproteinase-9) as a promising therapeutic target for GBM through bioinformatic predictions and human data analysis. Our results suggest that circMMP9 functions as a sponge for miR-149 and miR-542, both upregulated in GBM based on microarray data. Kaplan-Meier analysis indicated that reduced levels of miR-149 and miR-542 correlate with worse survival outcomes in GBM, suggesting their role as tumor suppressors. Importantly, miR-149 has been demonstrated to inhibit the expression of BIRC5 (baculoviral inhibitor of apoptosis repeat-containing 5 or survivin), a significant promoter of proliferation in GBM. BIRC5 is not only upregulated in GBM but also in various other cancers, including neuroblastoma and other brain cancers. Our protein-protein interaction analysis highlights the significance of BIRC5 as a central hub gene in GBM. CircMMP9 seems to influence this complex relationship by suppressing miR-149 and miR-542, despite their increased expression in GBM. Additionally, we found that circMMP9 directly interacts with heterogeneous nuclear ribonucleoproteins C and A1 (hnRNPC and A1), although not within their protein-binding domains. This suggests that hnRNPC/A1 may play a role in transporting circMMP9. Moreover, RNA-seq data from GBM patient samples confirmed the increased expression of BIRC5, PIK3CB, and hnRNPC/A1, further emphasizing the potential therapeutic significance of circMMP9 in GBM. In this study, we propose for the first time a new epigenetic regulatory role for circMMP9, highlighting a novel aspect of its oncogenic function. circMMP9 may regulate BIRC5 expression in GBM by sponging miR-149 and miR-542. BIRC5, in turn, suppresses apoptosis and enhances proliferation in GBM. Nonetheless, more extensive studies are advised to delve deeper into the roles of circMMP9, especially in the context of glioma.
Collapse
Affiliation(s)
- Javad Amini
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Nikta Zafarjafarzadeh
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sara Ghahramanlu
- Blood Transfusion Department of Samenolaemeh Hospital, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Omid Mohammadalizadeh
- Department of Agronomy and Plant Breeding, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Elaheh Mozaffari
- Biotechnology Research Center, Islamic Azad University of Shahrekord Branch, Tehran, Iran
| | - Bahram Bibak
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Nima Sanadgol
- Institute of Neuroanatomy, RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|
38
|
Yang JP, Kulkarni NN, Yamaji M, Shiraishi T, Pham T, Do H, Aiello N, Shaw M, Nakamura T, Abiru A, Gavva NR, Horman SR. Unveiling immune cell response disparities in human primary cancer-associated fibroblasts between two- and three-dimensional cultures. PLoS One 2024; 19:e0314227. [PMID: 39700125 DOI: 10.1371/journal.pone.0314227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/06/2024] [Indexed: 12/21/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) play pivotal roles in solid tumor initiation, growth, and immune evasion. However, the optimal biomimetic modeling conditions remain elusive. In this study, we investigated the effects of 2D and 3D culturing conditions on human primary CAFs integrated into a modular tumor microenvironment (TME). Using single-nucleus RNA sequencing (snRNAseq) and Proteomics' Proximity Extension Assays, we characterized CAF transcriptomic profiles and cytokine levels. Remarkably, when cultured in 2D, CAFs exhibited a myofibroblast (myCAF) subtype, whereas in 3D tumor spheroid cultures, CAFs displayed a more inflammatory (iCAF) pathological state. By integrating single-cell gene expression data with functional interrogations of critical TME-related processes [natural killer (NK)-mediated tumor killing, monocyte migration, and macrophage differentiation], we were able to reconcile form with function. In 3D TME spheroid models, CAFs enhance cancer cell growth and immunologically shield cells from NK cell-mediated cytotoxicity, in striking contrast with their 2D TME counterparts. Notably, 3D CAF-secreted proteins manifest a more immunosuppressive profile by enhancing monocyte transendothelial migration and differentiation into M2-like tumor-associated macrophages (TAMs). Our findings reveal a more immunosuppressive and clinically relevant desmoplastic TME model that can be employed in industrial drug discovery campaigns to expand the cellular target range of chemotherapeutics.
Collapse
Affiliation(s)
- Jian-Ping Yang
- Takeda Development Center Americas, Inc., San Diego, California, United States of America
| | - Nikhil Nitin Kulkarni
- Takeda Development Center Americas, Inc., San Diego, California, United States of America
| | - Masashi Yamaji
- Takeda Development Center Americas, Inc., San Diego, California, United States of America
| | | | - Thang Pham
- BioTuring, San Diego, California, United States of America
| | - Han Do
- BioTuring, San Diego, California, United States of America
| | - Nicole Aiello
- Bristol-Myers Squibb, Princeton, New Jersey, United States of America
| | - Michael Shaw
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts, United States of America
| | | | - Akiko Abiru
- Takeda Pharmaceutical Company Ltd, Fujisawa, Kanagawa, Japan
| | - Narender R Gavva
- Takeda Development Center Americas, Inc., San Diego, California, United States of America
| | - Shane R Horman
- Takeda Development Center Americas, Inc., San Diego, California, United States of America
| |
Collapse
|
39
|
Jędrzejewski T, Sobocińska J, Maciejewski B, Spisz P, Walczak-Skierska J, Pomastowski P, Wrotek S. In vitro treatment of triple-negative breast cancer cells with an extract from the Coriolus versicolor mushroom changes macrophage properties related to tumourigenesis. Immunol Res 2024; 73:14. [PMID: 39680299 DOI: 10.1007/s12026-024-09574-6] [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: 05/06/2024] [Accepted: 10/21/2024] [Indexed: 12/17/2024]
Abstract
Macrophages, the most abundant cells that participate in tumour progression, are the subject of a number of anticancer therapy approaches. Our previous results revealed that an extract of the fungus Coriolus versicolor (CV) has anti-cancer and immunomodulatory properties. The aim of the present study was to investigate whether CV extract-treated triple-negative breast cancer (TNBC) cells can release factors that can reprogram macrophages from pro-tumourigenic to anti-cancer subtypes. RAW 264.7 macrophages were cultured in a conditioned medium (CM) from non-treated 4T1 breast cancer cells (CM-NT) or CV extract-stimulated cells (CM-CV). After treatment, the following macrophage properties were evaluated: cell viability; M1/M2 phenotype (enzyme activities: iNOS and arginase 1; and expression of CD molecules: CD80 and CD163); cytokine concentrations: IL-6, TNF-α, IL-10, TGF-β, MCP-1 and VEGF; migration level; and ROS production. The results revealed that, compared with normal cells, TNBC cells stimulated with CV extract create a microenvironment that promotes a decrease in macrophage viability and migration, intracellular ROS production, and pro-angiogenic cytokine production (VEGF and MCP-1). Moreover, CM-CV decreased the expression of M2 macrophage markers (arginase 1 and CD163; IL-10 and TGF-β) but upregulated the expression of M1 cell markers (iNOS and CD80; IL-6 and TNF-α). We concluded that CV extract modifies the tumour microenvironment and changes macrophage polarisation toward functioning as an anti-tumour agent. Therefore, it is promising to use in the treatment of TNBC-associated macrophages.
Collapse
Affiliation(s)
- Tomasz Jędrzejewski
- Department of Immunology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Lwowska 1 Street, 87-100, Toruń, Poland.
| | - Justyna Sobocińska
- Department of Immunology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Lwowska 1 Street, 87-100, Toruń, Poland
| | - Bartosz Maciejewski
- Department of Immunology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Lwowska 1 Street, 87-100, Toruń, Poland
| | - Paulina Spisz
- Department of Immunology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Lwowska 1 Street, 87-100, Toruń, Poland
| | - Justyna Walczak-Skierska
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Toruń, Wileńska 4 Street, 87-100, Toruń, Poland
| | - Paweł Pomastowski
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Toruń, Wileńska 4 Street, 87-100, Toruń, Poland
- Department of Inorganic and Coordination Chemistry, Nicolaus Copernicus University in Toruń, Gagarina 7 Street, 87-100, Toruń, Poland
| | - Sylwia Wrotek
- Department of Immunology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Lwowska 1 Street, 87-100, Toruń, Poland
| |
Collapse
|
40
|
Anis A, Mostafa AM, Kerema MS, Hamdy NM, Sultan AS. In silico and cheminformatics prediction with experimental validation of an adipogenesis cocktail, sorafenib with rosiglitazone for HCC dedifferentiation. J Genet Eng Biotechnol 2024; 22:100429. [PMID: 39674644 PMCID: PMC11600669 DOI: 10.1016/j.jgeb.2024.100429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/06/2024] [Accepted: 10/07/2024] [Indexed: 12/16/2024]
Abstract
PURPOSE Hepatocellular carcinoma (HCC) resistance to sorafenib treatment and other treatment strategies causes a higher mortality rate in patients diagnosed with HCC. RESEARCH QUESTION HCC often develops resistance to sorafenib treatment and other therapies, leading to increased mortality rates in diagnosed patients. Herein, we propose a combined therapeutic approach using rosiglitazone, a key factor in cellular differentiation, along with adipogenesis inducers such as dexamethasone, IBMX, and insulin. Additionally, we included sorafenib, a primary drug for liver cancer treatment, in this combination cocktail and carried out the differentiation process in the presence of sorafenib. RESULTS Our study demonstrates that this combination induces the formation of adipocytes from HCC cells over several days under specific conditions and steps. CONCLUSION findings suggest that supplementing sorafenib with rosiglitazone and adipogenesis inducers may potentially transform HCC cells into adipocyte-like cells. Fat could be "the good" in the story of liver cancer alleviation, demonstrating the role of rosiglitazone.
Collapse
Affiliation(s)
- Aya Anis
- Postgraduate Pharmacist at DataClin CRO, Giza, Egypt
| | - Ahmed M Mostafa
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Abassia 11566, Cairo, Egypt
| | - Mariam S Kerema
- Postgraduate Pharmacist at Pharco Pharmaceuticals, Alexandria, Egypt
| | - Nadia M Hamdy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Abassia 11566, Cairo, Egypt.
| | - Ahmed S Sultan
- Biochemistry Department, Faculty of Science, Alexandria University, El-Shatbi, 21568, Alexandria, Egypt; Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States
| |
Collapse
|
41
|
Singhal S, Bhadana R, Jain BP, Gautam A, Pandey S, Rani V. Role of gut microbiota in tumorigenesis and antitumoral therapies: an updated review. Biotechnol Genet Eng Rev 2024; 40:3716-3742. [PMID: 36632709 DOI: 10.1080/02648725.2023.2166268] [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: 05/31/2022] [Indexed: 01/13/2023]
Abstract
Gut microbiota plays a prominent role in regulation of host nutrientmetabolism, drug and xenobiotics metabolism, immunomodulation and defense against pathogens. It synthesizes numerous metabolites thatmaintain the homeostasis of host. Any disbalance in the normalmicrobiota of gut can lead to pathological conditions includinginflammation and tumorigenesis. In the past few decades, theimportance of gut microbiota and its implication in various diseases, including cancer has been a prime focus in the field of research. Itplays a dual role in tumorigenesis, where it can accelerate as wellas inhibit the process. Various evidences validate the effects of gutmicrobiota in development and progression of malignancies, wheremanipulation of gut microbiota by probiotics, prebiotics, dietarymodifications and faecal microbiota transfer play a significant role.In this review, we focus on the current understanding of theinterrelationship between gut microbiota, immune system and cancer,the mechanisms by which they play dual role in promotion andinhibition of tumorigenesis. We have also discussed the role ofcertain bacteria with probiotic characteristics which can be used tomodulate the outcome of the various anti-cancer therapies under theinfluence of the alteration in the composition of gut microbiota.Future research primarily focusing on the microbiota as a communitywhich affect and modulate the treatment for cancer would benoteworthy in the field of oncology. This necessitates acomprehensive knowledge of the roles of individual as well asconsortium of microbiota in relation to physiology and response ofthe host.
Collapse
Affiliation(s)
- Shivani Singhal
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Renu Bhadana
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Buddhi Prakash Jain
- Department of Zoology, Mahatma Gandhi Central University, Motihari, Bihar, India
| | - Akash Gautam
- Centre for Neural and Cognitive Sciences, School of Medical Sciences, University of Hyderabad, Hyderabad, India
| | - Shweta Pandey
- Department of Biotechnology, Govt Vishwanath Yadav Tamaskar Post-Graduate Autonomous College Durg, Chhattisgarh, India
| | - Vibha Rani
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| |
Collapse
|
42
|
Yang H, Yang J, Zheng X, Chen T, Zhang R, Chen R, Cao T, Zeng F, Liu Q. The Hippo Pathway in Breast Cancer: The Extracellular Matrix and Hypoxia. Int J Mol Sci 2024; 25:12868. [PMID: 39684583 DOI: 10.3390/ijms252312868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
As one of the most prevalent malignant neoplasms among women globally, the optimization of therapeutic strategies for breast cancer has perpetually been a research hotspot. The tumor microenvironment (TME) is of paramount importance in the progression of breast cancer, among which the extracellular matrix (ECM) and hypoxia are two crucial factors. The alterations of these two factors are predominantly regulated by the Hippo signaling pathway, which promotes tumor invasiveness, metastasis, therapeutic resistance, and susceptibility. Hence, this review focuses on the Hippo pathway in breast cancer, specifically, how the ECM and hypoxia impact the biological traits and therapeutic responses of breast cancer. Moreover, the role of miRNAs in modulating ECM constituents was investigated, and hsa-miR-33b-3p was identified as a potential therapeutic target for breast cancer. The review provides theoretical foundations and potential therapeutic direction for clinical treatment strategies in breast cancer, with the aspiration of attaining more precise and effective treatment alternatives in the future.
Collapse
Affiliation(s)
- Hanyu Yang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Jiaxin Yang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Xiang Zheng
- School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Tianshun Chen
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Ranqi Zhang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Rui Chen
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Tingting Cao
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Fancai Zeng
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Qiuyu Liu
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
- Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| |
Collapse
|
43
|
Wang X, Wang J, Fu Q, Luo J, Shu M, Lin Z. Identification and interaction mechanism of novel small molecule antagonists targeting CC chemokine receptor 1/3/5 for treatment of non-small cell lung cancer. Mol Divers 2024:10.1007/s11030-024-11025-1. [PMID: 39578293 DOI: 10.1007/s11030-024-11025-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 10/18/2024] [Indexed: 11/24/2024]
Abstract
Non-Small Cell Lung Cancer (NSCLC) was one of the most prevalent forms of lung cancer. Due to its ease of invasion and migration, the five-year survival rate was relatively low. Therefore, new strategies for NSCLC treatment were needed. CC chemokine receptor 1/3/5 (CCR1/CCR3/CCR5), a member of the G-protein coupled receptor family, could promote the migration and invasion of NSCLC cells by binding to related chemokines. Consequently, targeting CCR1, CCR3 and CCR5 might prevent the progression of the disease. So far, no compound had been reported as a common antagonist for CCR1, CCR3, and CCR5. In this research, we utilized virtual screening and structural optimization to obtain compound 5, which effectively inhibited the migration and invasion of NSCLC cells. Meanwhile, Western Blot and Enzyme linked immunosorbent assay (ELISA) manifested that compound 5 suppressed migration and invasion of NSCLC cells by suppressing the nuclear factor κB (NF-κB) and the consequently decreased Matrix Metalloproteinase-9(MMP-9) secretion. Moreover, drug affinity responsive target stability (DARTS) experiment and molecular simulations confirmed that compound 5 was capable of binding with CCR1/CCR3/CCR5, and Van der Waals forces were instrumental in the binding process. Ile91, Tyr113, Gln284, and Ser184(CCR1-ligand5), Ile189, Met213, and Leu209(CCR3-ligand5), Phe109, Gln194, and Thr195(CCR5-ligand5) had Van der Waals interactions with ligand 5. Dynamic cross-correlation matrix (DCCM) and free energy landscape (FEL) showed that compound 5 could stably bind to CCR1/CCR3/CCR5 to change conformation of the protein and the tendency of residue movements, leading to a persistent inhibitory effect. This study aimed to provide assistance in the rational design of common antagonists for CCR1, CCR3, and CCR5.
Collapse
Affiliation(s)
- Xiaomeng Wang
- Department of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
- Key Laboratory of Screening and Activity Evaluation of Targeted Drugs, Chongqing, 400054, China
| | - Juan Wang
- Department of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China.
- Key Laboratory of Screening and Activity Evaluation of Targeted Drugs, Chongqing, 400054, China.
- Department of Pathophysiology, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China.
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, 400038, People's Republic of China.
- Key Laboratory of High Altitude Medicine, PLA, Chongqing, 400038, People's Republic of China.
| | - Qiao Fu
- Department of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
- Key Laboratory of Screening and Activity Evaluation of Targeted Drugs, Chongqing, 400054, China
| | - Jing Luo
- Department of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
- Key Laboratory of Screening and Activity Evaluation of Targeted Drugs, Chongqing, 400054, China
| | - Mao Shu
- Department of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
- Key Laboratory of Screening and Activity Evaluation of Targeted Drugs, Chongqing, 400054, China
| | - Zhihua Lin
- Chongqing College of Traditional Chinese Medicine, Chongqing, 402760, China.
| |
Collapse
|
44
|
Yao Y, Shen G, Luo J, Wang J, Xu Z, Wang H, Cui L. Research Progress with Atractylone as an Antitumor Agent. Molecules 2024; 29:5450. [PMID: 39598839 PMCID: PMC11597220 DOI: 10.3390/molecules29225450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/30/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
Atractylone is a sesquiterpenoid compound extracted from Rhizoma Atractylodis. As one of the main active components in the volatile oil of the Atractylodes genus, it has exhibited certain therapeutic effects, including anti-inflammatory, antiviral, antioxidant, antiallergic, antiangiogenic, and neuroprotective activities, among others. With further research on the chemical constituents and pharmacology of sesquiterpenes, research on the antitumor activity of Atractylone has also been further expanded. Much of the current literature pays particular attention to the antitumor activity of Atractylone, which was found to inhibit the apoptosis of tumor cells and prevent growth, invasion, and migration through different apoptosis pathways and signaling pathways. Due to its promising potential for cancer prevention, it may play a role in reducing the incidence of malignant tumors. In this paper, the antitumor activity and mechanism of Atractylone are reviewed, providing a reference to inform future research on the tumor treatment, clinical application, and further development and utilization of this plant genus.
Collapse
Affiliation(s)
- Ying Yao
- College of Pharmacy, Harbin University of Commerce, Harbin 150076, China; (Y.Y.); (G.S.); (J.L.); (J.W.); (Z.X.)
- Heilongjiang Provincial Key Laboratory of Geriatric Medicine, Harbin 150076, China
| | - Guanghuan Shen
- College of Pharmacy, Harbin University of Commerce, Harbin 150076, China; (Y.Y.); (G.S.); (J.L.); (J.W.); (Z.X.)
- Heilongjiang Provincial Key Laboratory of Geriatric Medicine, Harbin 150076, China
- Postdoctoral Programme of Meteria Medica Institute, Harbin University of Commerce, Harbin 150076, China
| | - Jianghan Luo
- College of Pharmacy, Harbin University of Commerce, Harbin 150076, China; (Y.Y.); (G.S.); (J.L.); (J.W.); (Z.X.)
- Heilongjiang Provincial Key Laboratory of Geriatric Medicine, Harbin 150076, China
| | - Jinhong Wang
- College of Pharmacy, Harbin University of Commerce, Harbin 150076, China; (Y.Y.); (G.S.); (J.L.); (J.W.); (Z.X.)
- Heilongjiang Provincial Key Laboratory of Geriatric Medicine, Harbin 150076, China
| | - Zheng Xu
- College of Pharmacy, Harbin University of Commerce, Harbin 150076, China; (Y.Y.); (G.S.); (J.L.); (J.W.); (Z.X.)
- Heilongjiang Provincial Key Laboratory of Geriatric Medicine, Harbin 150076, China
| | - Hao Wang
- College of Pharmacy, Harbin University of Commerce, Harbin 150076, China; (Y.Y.); (G.S.); (J.L.); (J.W.); (Z.X.)
- Heilongjiang Provincial Key Laboratory of Geriatric Medicine, Harbin 150076, China
| | - Linlin Cui
- College of Pharmacy, Harbin University of Commerce, Harbin 150076, China; (Y.Y.); (G.S.); (J.L.); (J.W.); (Z.X.)
- Heilongjiang Provincial Key Laboratory of Geriatric Medicine, Harbin 150076, China
| |
Collapse
|
45
|
Mistretta KS, Coburn JM. Three-dimensional silk fibroin scaffolded co-culture of human neuroblastoma and innate immune cells. Exp Cell Res 2024; 443:114289. [PMID: 39433171 DOI: 10.1016/j.yexcr.2024.114289] [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: 06/17/2024] [Revised: 09/18/2024] [Accepted: 10/18/2024] [Indexed: 10/23/2024]
Abstract
Neuroblastoma (NB) is the most common pediatric extracranial solid tumor. It accounts for 50 % of cancers diagnosed in infants less than 1 year old, and 10 % of all pediatric cancer deaths in the United States. High-risk patients have a less than 50 % 5-year survival rate with current treatment strategies. The complex tumor microenvironment of NB makes the development of treatment strategies for high-risk patients challenging. There is increasing evidence that intratumoral immune suppression plays an important role in the progression and invasion of NB tumors. Few three-dimensional (3D) cancer models include components of the innate immune system. This work develops a preclinical 3D NB-immune co-culture model using SK-N-AS NB cells, NK-92 natural killer cells, and THP-1 derived macrophages, co-cultured on porous 3D silk scaffolds to provide tumor architecture. Conditioned media and indirect co-culturing showed changes in SK-N-AS gene expression associated with immunoregulatory signaling, and changes in NK-92 gene expression that are associated with reduced cytotoxicity. This motivated the development of a 3D direct co-culture system in which NB cells were seeded prior to immune cells to allow incorporation and deposition of extracellular matrix within the construct. Immune cells were then incorporated into the model to achieve direct co-culture with SK-N-AS cells. Changes in THP-1 macrophage polarization toward a more M2-like phenotype were observed in 3D direct co-culture, as well as altered NK-92 cell protein secretion and cytotoxic activity. Preliminary testing of immunotherapeutics within the model was conducted on both NB-macrophage and NB-NK co-cultures, but the model demonstrated limited response to immunotherapeutics. This work lays the foundation for building high-throughput therapeutic screening models for the improved treatment NB and other solid tumors.
Collapse
Affiliation(s)
- Katelyn S Mistretta
- Department of Biomedical Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA
| | - Jeannine M Coburn
- Department of Biomedical Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA.
| |
Collapse
|
46
|
Yang GR, Kim W, Jung JH. Sliding Microneedle - Lateral flow immunoassay strip device for highly sensitive biomarker detection in interstitial fluid. Biosens Bioelectron 2024; 263:116590. [PMID: 39096764 DOI: 10.1016/j.bios.2024.116590] [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: 06/07/2024] [Revised: 07/11/2024] [Accepted: 07/16/2024] [Indexed: 08/05/2024]
Abstract
Diabetes is a chronic disease with significant complications, necessitating regular treatment and checkups, which can be costly and time-consuming for patients. To address this, we developed the Sliding Microneedle (MN)-Lateral flow immunoassay strip (LFIAs) device that combines the advantages of MNs and LFIAs to detect IL-6, an independent biomarker for diabetes complications. This device offers rapid and highly sensitive detection of IL-6 by extracting interstitial fluid (ISF) through MNs and transferring it to LFIAs. The stainless MN, embedded in the 3D-printed Sliding MN-LFIAs device, was inserted into the skin at a 20° angle, minimizing blood contamination risk. With a filter paper attached to the MN surface, the device collected 4.65 ± 0.05 μL of ISF containing IL-6 within 90 s. The ISF was then transferred to the LFIAs using a running buffer. After a 15-min reaction, silver enhancement (SE) treatment was applied, allowing for the highly sensitive and specific detection of IL-6 at 102 pg/mL concentrations. The Sliding MN-LFIAs device successfully distinguished between normal and diabetic rat models, demonstrating its potential as an effective tool for detecting diabetes complications quickly and affordably.
Collapse
Affiliation(s)
- Ga Ram Yang
- Department of Pharmaceutical Engineering, Dankook University, Cheonan, South Korea
| | - Woojin Kim
- Department of Pharmaceutical Engineering, Dankook University, Cheonan, South Korea
| | - Jae Hwan Jung
- Department of Pharmaceutical Engineering, Dankook University, Cheonan, South Korea.
| |
Collapse
|
47
|
Guo M, Sheng W, Yuan X, Wang X. Neutrophils as promising therapeutic targets in pancreatic cancer liver metastasis. Int Immunopharmacol 2024; 140:112888. [PMID: 39133956 DOI: 10.1016/j.intimp.2024.112888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/01/2024] [Accepted: 08/04/2024] [Indexed: 09/01/2024]
Abstract
Pancreatic cancer is characterized by an extremely poor prognosis and presents significant treatment challenges. Liver metastasis is the leading cause of death in patients with pancreatic cancer. Recent studies have highlighted the significant impact of neutrophils on tumor occurrence and progression, as well as their crucial role in the pancreatic cancer tumor microenvironment. Neutrophil infiltration plays a critical role in the progression and prognosis of pancreatic cancer. Neutrophils contribute to pancreatic cancer liver metastasis through various mechanisms, including angiogenesis, immune suppression, immune evasion, and epithelial-mesenchymal transition (EMT). Therefore, targeting neutrophils holds promise as an important therapeutic strategy for inhibiting pancreatic cancer liver metastasis. This article provides a summary of research findings on the involvement of neutrophils in pancreatic cancer liver metastasis and analyzes their potential as therapeutic targets. This research may provide new insights for the treatment of pancreatic cancer and improve the prognosis of patients with this disease.
Collapse
Affiliation(s)
- Minjie Guo
- Department of Thoracic Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Wanying Sheng
- Department of Thoracic Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiao Yuan
- Cancer Institute of Jiangsu University, Zhenjiang, China.
| | - Xu Wang
- Department of Thoracic Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| |
Collapse
|
48
|
Najafzadeh M, Sajjadi SM, Kharazi S, Karimifard F, Safarpour H, Kharazinejad E. Interactions between cancer and stroma mediated by extracellular vesicles. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2024; 25:114. [DOI: 10.1186/s43042-024-00582-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 09/13/2024] [Indexed: 01/05/2025] Open
Abstract
AbstractExtracellular vehicles (EVs) are small membrane-bound particles that are released by both cancer and stromal cells. These vesicles have emerged as key mediators of intercellular communication within the tumor microenvironment. In particular, EVs have been shown to play a critical role in facilitating the interactions between cancer cells and the surrounding stroma. Through the transfer of various bioactive molecules, including proteins, lipids, and nucleic acids, EVs are able to modulate the behavior of recipient cells and promote tumorigenesis. Additionally, EVs can also contribute to the development of drug resistance and immune evasion, further highlighting their importance in cancer progression. This review will summarize the current knowledge regarding EV-mediated interactions between cancer and stromal cells, and discuss their implications for cancer diagnosis and therapy.
Collapse
|
49
|
Pang G, Ye L, Jiang Y, Wu Y, Zhang R, Yang H, Yang Y. Unveiling the bidirectional role of MMP9: A key player in kidney injury. Cell Signal 2024; 122:111312. [PMID: 39074714 DOI: 10.1016/j.cellsig.2024.111312] [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: 05/15/2024] [Revised: 07/04/2024] [Accepted: 07/22/2024] [Indexed: 07/31/2024]
Abstract
Matrix metalloproteinases (MMPs) are a group of zinc-dependent proteolytic metalloenzymes that are involved in numerous pathological conditions, including nephropathy. MMP9, a member of the MMPs family, is categorized as a constituent of the gelatinase B subgroup, and its involvement in extracellular matrix (ECM) remodeling and renal fibrosis highlights its importance in the development and progression of renal diseases. The exact role of MMP9 in the development of kidney diseases is still controversial. This study investigated the dual role of MMP9 in kidney injury, discussing its implications in the pathogenesis of kidney diseases and investigating the design and mechanism of MMP9 inhibitors based on previous studies. This study provides an effective basis for the development of novel and selective MMP9 inhibitors for treating renal diseases.
Collapse
Affiliation(s)
- Guiying Pang
- Anhui University of Traditional Chinese Medicine, Hefei 230000, People's Republic of China; Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd., Beijing 102609, People's Republic of China; Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong 226133, People's Republic of China
| | - Ling Ye
- Anhui University of Traditional Chinese Medicine, Hefei 230000, People's Republic of China; Department of Pharmacology, Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing 102609, People's Republic of China; Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong 226133, People's Republic of China
| | - Yinxiao Jiang
- Anhui University of Traditional Chinese Medicine, Hefei 230000, People's Republic of China; Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong 226133, People's Republic of China
| | - Yilin Wu
- Anhui University of Traditional Chinese Medicine, Hefei 230000, People's Republic of China; Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd., Beijing 102609, People's Republic of China; Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong 226133, People's Republic of China
| | - Rufeng Zhang
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd., Beijing 102609, People's Republic of China; Department of Pharmacology, Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing 102609, People's Republic of China
| | - Hongxu Yang
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd., Beijing 102609, People's Republic of China.
| | - Yi Yang
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd., Beijing 102609, People's Republic of China; Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong 226133, People's Republic of China.
| |
Collapse
|
50
|
Zhu J, Mei J, He Y, Zou Y, Hu X. Chlorogenic acid exhibits antitumor effect in patient-derived xenograft models and hydrogel-embedded tissue culture drug susceptibility test of tongue cancer. Heliyon 2024; 10:e37523. [PMID: 39309775 PMCID: PMC11416273 DOI: 10.1016/j.heliyon.2024.e37523] [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: 04/08/2024] [Revised: 08/03/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024] Open
Abstract
Chlorogenic acid (CGA) is one of the effective components of Chinese medicine plant such as honeysuckle and Eucommia ulmoides. CGA can inhibits various cancer types, but its effectivity against tongue cancer remains unknown. In the present study, we utilized patient-derived xenograft (PDX) models in conjunction with hydrogel-embedded drug sensitivity tests (HDST) to demonstrate the inhibitory effects of CGA on tongue cancer tissues in both in vivo and ex vivo experimental paradigms. Immunohistochemical (IHC) analysis and TUNEL staining revealed that CGA downregulated the expression of CD31 and Ki-67, while concurrently promoting apoptosis. Furthermore, the involvement of the EGFR-AKT-MMP9 signaling cascade in the tumor-suppressive effects of CGA was confirmed using network pharmacology analysis and immunofluorescent validation techniques. Overall, our findings indicate that CGA robustly inhibits tongue cancer in cellular and organismal models. The EGFR-AKT-MMP9 axis plays a highly significant role in mediating this bioactivity, thereby positioning CGA as a promising candidate for further investigation in oncology. The multifaceted therapeutic potential of CGA, as evidenced by its ability to disrupt angiogenesis, suppress cell proliferation, and induce apoptosis, underscores its value as a novel therapeutic agent for the treatment of tongue cancer.
Collapse
Affiliation(s)
- Jia Zhu
- The Affiliated Stomatological Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Oral Diseases, Nanchang, Jiangxi, China
- Jiangxi Provincial Clinical Research Center for Oral Diseases, Nanchang, Jiangxi, China
| | - Jiaqi Mei
- Department of Hematology, University Hospitals and University of the Second Clinical Medical College of Nanchang University, Nanchang, Jiangxi, China
| | - Yuanqiao He
- Nanchang Royo Biotech Co., Ltd, Nanchang, Jiangxi, China
- Center of Laboratory Animal Science, Nanchang University, Nanchang, Jiangxi, China
| | - Yan Zou
- The Affiliated Stomatological Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Oral Diseases, Nanchang, Jiangxi, China
- Jiangxi Provincial Clinical Research Center for Oral Diseases, Nanchang, Jiangxi, China
| | - Xiaoping Hu
- The Affiliated Stomatological Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Oral Diseases, Nanchang, Jiangxi, China
- Jiangxi Provincial Clinical Research Center for Oral Diseases, Nanchang, Jiangxi, China
| |
Collapse
|