1
|
He L, Jiang Z, Wang J, Han Z. Mechanism of miR-200b-3p-induced FOSL2 inhibition of endometrial cancer cell proliferation and metastasis. Sci Rep 2025; 15:15742. [PMID: 40325054 PMCID: PMC12052843 DOI: 10.1038/s41598-025-00224-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: 08/06/2024] [Accepted: 04/25/2025] [Indexed: 05/07/2025] Open
Abstract
The purpose of this study was to investigate how miR-200b-3p inhibits the proliferation and metastasis of endometrial cancer cells by inducing the expression of FOSL2 in the AP1 transcription family. Endometrial cancer cell line HEC-1-A was divided into 16 groups: NC-mimic (transfected with negative control NC mimic), miR-200b-3p mimic (transfected with miR-200b-3p mimic), NC-suppress (transfected with negative control NC inhibit), miR-200b-3p inhibit group (transfected with miR-200b-3p inhibit), si-NC (transfected with negative control si-NC), si-FOSL2 (transfected with Si-FOSL2), oe-NC (transfected with negative control oe-NC), oe-FOSL2 group (oe-FOSL2), miR-200b-3p mimic + oe-NC group (co-transfected with miR-200b-3p mimic and oe-NC), miR-200b-3p mimic + oe-FOSL2 group (co-transfected with miR-200b-3p mimic and oe-FOSL2), miR-200b-3p inhibit + si-NC group (co-transfected with miR-200b-3p inhibit and si-NC), miR-200b-3p inhibit + si-FOSL2 group (co-transfected with miR-200b-3p inhibit and si-FOSL2), miR-200b-3p mimic + si-NC group (co-transfected with miR-200b-3p mimic and si-NC), miR-200b-3p mimic + si-FOSL2 group (co-transfected with miR-200b-3p mimic and si-FOSL2), miR-200b-3p inhibit + oe-NC group (co-transfected with miR-200b-3p inhibit and oe-NC), miR-200b-3p inhibit + oe-FOSL2 group (co-transfected with miR-200b-3p inhibit and oe-FOSL2). Real-time fluorescence quantitative PCR, Western blot, CCK-8 assay, scratch test and Transwell assay were used to detect the expression of miR-200b-3p mRNA, FOSL2 mRNA and protein, cell proliferation, migration and invasion. In endometrial cancer cell lines, the expression of miR-200b-3p was significantly down-regulated (P < 0.05), while the expression of FOSL2 was significantly up-regulated (P < 0.05). Compared with NC-mimic group, the expression of FOSL2, N-cadherin and Vimentin in miR-200b-3p mimic group was significantly decreased (P < 0.05), and the expression of E-cadherin was significantly increased (P < 0.05). The cell proliferation, migration rate and the number of transmembrane cells were significantly decreased (P < 0.05). Compared with the miR-200b-3p mimic + oe-NC group, the expression of FOSL2, N-cadherin and Vimentin in miR-200b-3p mimic + oe-FOSL2 was significantly increased (P < 0.05), the expression level of E-cadherin was significantly decreased (P < 0.05), and the cell proliferation, migration rate and the number of transmembrane cells were significantly increased (P < 0.05). Compared with NC-inhibit group, the expression of FOSL2, N-cadherin and Vimentin in miR-200b-3p inhibit group was significantly increased (P < 0.05), and the expression of E-cadherin was significantly decreased (P < 0.05). The cell proliferation, migration rate and the number of transmembrane cells were significantly increased (P < 0.05). Compared with the miR-200b-3p inhibit + si-NC group, the expression of FOSL2, N-cadherin and Vimentin in miR-200b-3p inhibit + si-FOSL2 was significantly decreased (P < 0.05), and the expression of E-cadherin was significantly increased (P < 0.05); the cell proliferation, migration rate and the number of transmembrane cells were significantly decreased (P < 0.05) The expression of miR-200b-3p in endometrial cancer cells is down-regulated, which can inhibit the proliferation, migration and invasion of endometrial cancer cells by regulating the EMT process, and its mechanism is related to its targeted negative regulation of FOSL2 expression.
Collapse
Affiliation(s)
- Lijie He
- Department of Clinical Laboratory, Tianjin Fifth Central Hospital, 41 Zhejiang Road, Tianjin, 300450, People's Republic of China
| | - Zhongmin Jiang
- Department of Pathology, Tianjin Fifth Central Hospital, Tianjin, 300450, People's Republic of China
| | - Jing Wang
- Department of Clinical Laboratory, Tianjin Fifth Central Hospital, 41 Zhejiang Road, Tianjin, 300450, People's Republic of China
| | - Zhe Han
- Department of Clinical Laboratory, Tianjin Fifth Central Hospital, 41 Zhejiang Road, Tianjin, 300450, People's Republic of China.
| |
Collapse
|
2
|
Ghafoor S, Garcia E, Jay DJ, Persad S. Molecular Mechanisms Regulating Epithelial Mesenchymal Transition (EMT) to Promote Cancer Progression. Int J Mol Sci 2025; 26:4364. [PMID: 40362600 PMCID: PMC12072817 DOI: 10.3390/ijms26094364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
The process of epithelial-mesenchymal transition (EMT) is crucial in various physiological/pathological circumstances such as development, wound healing, stem cell behavior, and cancer progression. It involves the conversion of epithelial cells into a mesenchymal phenotype, which causes the cells to become highly motile. This reprogramming is initiated and controlled by various signaling pathways and governed by several key transcription factors, including Snail 1, Snail 2 (Slug), TWIST 1, TWIST2, ZEB1, ZEB2, PRRX1, GOOSECOID, E47, FOXC2, SOX4, SOX9, HAND1, and HAND2. The intracellular signaling pathways are activated/inactivated by signals received from the extracellular environment and the transcription factors are carefully regulated at the transcriptional, translational, and post-translational levels to maintain tight regulatory control of EMT. One of the most important pathways involved in this process is the transforming growth factor-β (TGFβ) family signaling pathway. This review will discuss the role of EMT in promoting epithelial cancer progression and the convergence/interplay of multiple signaling pathways and transcription factors that regulate this phenomenon.
Collapse
Affiliation(s)
| | | | | | - Sujata Persad
- Department of Pediatrics, Faculty of Medicine and Dentistry, 3020R Katz Group Centre for Pharmacy and Health Research, University of Alberta, Edmonton, AB T6G 2E1, Canada; (S.G.); (E.G.); (D.J.J.)
| |
Collapse
|
3
|
Fu Y, Qin C, Li M, Zhang X, Gai Y, Ruan W, Lan X. Comparative Evaluation of 68Ga-FAPI-04 PET for Initial N and M Staging in Gastric Cancer: A Study Against Histopathology and Contrast-Enhanced CT. Clin Nucl Med 2025; 50:394-403. [PMID: 40179292 DOI: 10.1097/rlu.0000000000005795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 01/23/2025] [Indexed: 04/05/2025]
Abstract
PURPOSE To evaluate the efficiency of 68Ga-FAPI-04 PET (PET/MRI or PET/CT) for N and M staging in gastric carcinoma and compare outcomes with histopathology and contrast-enhanced computed tomography (CECT). PATIENTS AND METHODS Patients with gastric carcinoma who had undergone 68Ga-FAPI-04 PET/MRI or PET/CT before treatment were retrospectively enrolled. Histopathology post lymphadenectomy was the gold standard for N staging, while histopathology and follow-up data were the reference for overall outcomes. The diagnostic efficiency of 68Ga-FAPI-04 PET for detecting regional lymph node involvement and distant metastases was compared to that of CECT. RESULTS Sixty-two patients were enrolled. In 18 patients who underwent 68Ga-FAPI-04 PET/MRI and lymphadenectomy, 532 lymph nodes were dissected. 68Ga-FAPI-04 PET/MRI showed similar sensitivity, specificity, and accuracy compared to CECT (28.3% vs. 23.2%, 99.8% vs. 99.3%, and 86.5% vs. 85.2%, all P > 0.05). Fifty-five patients had regional lymph node metastasis, 68Ga-FAPI-04 PET exhibited comparable diagnostic efficiency to CECT, with sensitivity of 83.6% versus 87.3%, specificity of 100% versus 85.7%, accuracy of 85.5% versus 87.1% (all P > 0.05). Excluding 3 patients with only abdominal CECT, 32 out of 59 patients had distant metastasis, with no significant differences in sensitivity, specificity, and accuracy between 68Ga-FAPI-04 PET and CECT based on patient (100% vs. 87.5%, 92.6% vs. 96.3%, and 96.6% vs. 91.5%, all P >0.05). Notably, 68Ga-FAPI-04 PET outperformed CECT in detecting peritoneal, distant lymph nodes, bone, liver, and ovarian metastases by visualizing more lesions or greater lesion extent. CONCLUSIONS 68Ga-FAPI-04 PET exhibits comparable diagnostic performance to CECT for patient-based N staging and M staging of gastric cancer. However, it surpasses CECT in visualizing distant metastases.
Collapse
Affiliation(s)
- Yiru Fu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Chunxia Qin
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Mengting Li
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Xiao Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yongkang Gai
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Weiwei Ruan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| |
Collapse
|
4
|
Feigelman G, Simanovich E, Rahat MA. Serum EMMPRIN/CD147 promotes the lung pre-metastatic niche in a D2A1 mammary carcinoma mouse model. Front Immunol 2025; 16:1568578. [PMID: 40370456 PMCID: PMC12075191 DOI: 10.3389/fimmu.2025.1568578] [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: 01/30/2025] [Accepted: 04/08/2025] [Indexed: 05/16/2025] Open
Abstract
Several types of cancer, including breast cancer, metastasize to the lung. However, before the disseminating tumor cells (DTCs) arrive there, the lung must be prepared as a hospitable environment for them, forming the pre-metastatic niche (PMN). It is now accepted that the primary tumor can release soluble mediators or extracellular vesicles that activate the PMN resident cells, recruit immune cells, promote angiogenesis, and remodel the extracellular matrix (ECM), even before the arrival of DTCs to the niche. However, not all the components of the tumor secretome are known. Here we demonstrate in a mouse model of breast cancer designed to generate lung PMN, that EMMPRIN, a multifunction protein and mediator of tumor-stroma cell interactions, is part of that secretome. To study the involvement of EMMPRIN in the generation of lung PMN, we knocked down its expression in D2A1 cells (D2A1-KD), treated the mice with the anti-EMMPRIN antibody developed in our lab (m161-pAb), or administered the recombinant EMMPRIN protein to healthy mice. We show that the primary tumor released elevated levels of EMMPRIN in mice implanted with paternal D2A1 cells (D2A1-WT), generating lung PMN by increasing VEGF, MMP-9 and TGFβ secretion, enhancing angiogenesis, activating fibroblasts, increasing neutrophils infiltration, and remodeling the ECM. These effects were inhibited in mice implanted with D2A1-KD cells or administered with m161-pAb. In healthy mice, the recombinant EMMRPIN recapitulated the effects of high EMMPRIN levels. Thus, EMMPRIN as part of the tumor secretome is sufficient to promote the lung PMN, and targeting it could potentially inhibit the metastatic cascade.
Collapse
Affiliation(s)
- Gabriele Feigelman
- Immunotherapy Laboratory, Research Laboratories, Carmel Medical Center, Haifa, Israel
| | - Elina Simanovich
- Immunotherapy Laboratory, Research Laboratories, Carmel Medical Center, Haifa, Israel
| | - Michal A. Rahat
- Immunotherapy Laboratory, Research Laboratories, Carmel Medical Center, Haifa, Israel
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
5
|
Liang Z, Li S, Wang Z, Zhou J, Huang Z, Li J, Bao H, Yam JWP, Xu Y. Unraveling the Role of the Wnt Pathway in Hepatocellular Carcinoma: From Molecular Mechanisms to Therapeutic Implications. J Clin Transl Hepatol 2025; 13:315-326. [PMID: 40206274 PMCID: PMC11976435 DOI: 10.14218/jcth.2024.00401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 04/11/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the deadliest malignant tumors in the world, and its incidence and mortality have increased year by year. HCC research has increasingly focused on understanding its pathogenesis and developing treatments.The Wnt signaling pathway, a complex and evolutionarily conserved signal transduction system, has been extensively studied in the genesis and treatment of several malignant tumors. Recent investigations suggest that the pathogenesis of HCC may be significantly influenced by dysregulated Wnt/β-catenin signaling. This article aimed to examine the pathway that controls Wnt signaling in HCC and its mechanisms. In addition, we highlighted the role of this pathway in HCC etiology and targeted treatment.
Collapse
Affiliation(s)
- Zixin Liang
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shanshan Li
- School of Pharmacy, Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu Medical University, Bengbu, Anhui, China
| | - Zhiyu Wang
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Junting Zhou
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ziyue Huang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Jiehan Li
- Department of Urology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Haolin Bao
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Judy Wai Ping Yam
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yi Xu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- School of Pharmacy, Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu Medical University, Bengbu, Anhui, China
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
6
|
Cao L, Leclercq-Cohen G, Klein C, Sorrentino A, Bacac M. Mechanistic insights into resistance mechanisms to T cell engagers. Front Immunol 2025; 16:1583044. [PMID: 40330489 PMCID: PMC12053166 DOI: 10.3389/fimmu.2025.1583044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 03/31/2025] [Indexed: 05/08/2025] Open
Abstract
T cell engagers (TCEs) represent a groundbreaking advancement in the treatment of B and plasma cell malignancies and are emerging as a promising therapeutic approach for the treatment of solid tumors. These molecules harness T cells to bind to and eliminate cancer cells, effectively bypassing the need for antigen-specific T cell recognition. Despite their established clinical efficacy, a subset of patients is either refractory to TCE treatment (e.g. primary resistance) or develops resistance during the course of TCE therapy (e.g. acquired or treatment-induced resistance). In this review we comprehensively describe the resistance mechanisms to TCEs, occurring in both preclinical models and clinical trials with a particular emphasis on cellular and molecular pathways underlying the resistance process. We classify these mechanisms into tumor intrinsic and tumor extrinsic ones. Tumor intrinsic mechanisms encompass changes within tumor cells that impact the T cell-mediated cytotoxicity, including tumor antigen loss, the expression of immune checkpoint inhibitory ligands and intracellular pathways that render tumor cells resistant to killing. Tumor extrinsic mechanisms involve factors external to tumor cells, including the presence of an immunosuppressive tumor microenvironment (TME) and reduced T cell functionality. We further propose actionable strategies to overcome resistance offering potential avenues for enhancing TCE efficacy in the clinic.
Collapse
Affiliation(s)
- Linlin Cao
- Roche Innovation Center, Zürich, Switzerland
| | | | | | | | | |
Collapse
|
7
|
Ba J, Zheng C, Lai Y, He X, Pan Y, Zhao Y, Xie H, Wu B, Deng X, Wang N. High matrix stiffness promotes senescence of type II alveolar epithelial cells by lysosomal degradation of lamin A/C in pulmonary fibrosis. Respir Res 2025; 26:128. [PMID: 40205454 PMCID: PMC11984030 DOI: 10.1186/s12931-025-03201-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 03/24/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Cellular senescence is one of the key steps in the progression of pulmonary fibrosis, and the senescence of type II alveolar epithelial cells (AEC IIs) may potentially accelerate the progression of pulmonary fibrosis. However, the molecular mechanisms underlying cellular senescence in pulmonary fibrosis remain unclear. METHODS The researchers first conducted in vitro experiments to investigate whether AEC IIs cultured on high matrix stiffness would lead to cellular senescence. Next, samples from mouse pulmonary fibrosis models and clinical idiopathic pulmonary fibrosis (IPF) patients were tested to observe extracellular matrix deposition, lamin A/C levels, and cellular senescence status in lung tissue. Construct lamin A/C knockdown and overexpression systems separately in AEC IIs, and observe whether changes in lamin A/C levels lead to cellular senescence. Further explore the degradation mechanism of lamin A/C using protein degradation inhibitors. RESULTS In vitro experiments have found that high matrix stiffness promotes senescence of AEC IIs. In a mouse model of pulmonary fibrosis, AEC IIs were found to exhibit significant cellular senescence on day 21. In clinical IPF samples, it was found that senescent cells expressed low levels of lamin A/C. In the lamin A/C SiRNA knockdown system, it was further confirmed that AEC IIs with low levels of lamin A/C are more prone to cellular senescence. Under high matrix stiffness, lamin A/C in AEC IIs is degraded through the autophagy lysosome pathway. The use of chloroquine can effectively alleviate cellular senescence. CONCLUSIONS High matrix stiffness degrades lamin A/C in pulmonary fibrosis through lysosomal degradation pathways, promoting AEC II senescence. Inhibition the degradation of lamin A/C could alleviate AEC II senescence.
Collapse
Affiliation(s)
- Junhui Ba
- Department of Medical Intensive Care Unit, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Changyu Zheng
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Yimei Lai
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Xin He
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Yuxi Pan
- Department of Oncology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Yanqiu Zhao
- Shenzhen Samii Medical Center, Shenzhen, Guangdong Province, China
| | - Huihui Xie
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Benquan Wu
- Department of Medical Intensive Care Unit, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China.
| | - Xiao Deng
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China.
| | - Nan Wang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, China.
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong Province, China.
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, China.
| |
Collapse
|
8
|
Wei Y, Wei S, Lei Z, Zhang Y, Wu J, Huang J, Fu L, Li Z, Huang G, Liang Y, Zheng J. USP4 promotes proliferation and metastasis in human lung adenocarcinoma. Sci Rep 2025; 15:11096. [PMID: 40169699 PMCID: PMC11961685 DOI: 10.1038/s41598-025-89377-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 02/05/2025] [Indexed: 04/03/2025] Open
Abstract
Research the expression of USP4 in lung adenocarcinoma and its correlation with clinicopathological features and prognosis analysis, to explore the invasion and metastasis mechanism of USP4 in lung adenocarcinoma, and to clarify the mechanism of USP4's involvement in the occurrence and development of lung adenocarcinoma. The expressions of USP4, VEGF, MMP2 and Ki67 in lung adenocarcinoma and adjacent tissues of 139 patients with lung adenocarcinoma were detected by immunohistochemical method, and the correlation between expression and clinicopathological features and survival curve were analyzed by statistical method. The expression of USP4 was interfered by LIP-2000 cell transfection technology, and the expression of USP4 and its related factors in protein level was detected by Western Blot, and their correlation was analyzed. After silencing USP4 expression, the effects of USP4 on proliferation, invasion and migration of lung adenocarcinoma cells were detected by cell scratches assay, MTT assay, Transwell assay and tumorigenesis assay in nude mice. The expression of USP4 in lung adenocarcinoma tissues was higher than that in normal adjacent tissues, and the high expression of USP4 was significantly correlated with the differentiation degree of lung adenocarcinoma, clinical stage and pathological grade lymph node metastasis. After silencing USP4 expression, the expression of cyclin apoptosis protein invasion related proteins and phosphorylation factors were affected, and then cell migration and the proliferation ability decreased, the number of invasion and metastasis decreased, and the tumor volume decreased in nude mice. USP4 may play a certain role in the invasion and metastasis of lung adenocarcinoma by regulating the expression of tumor-related factors and affecting the prognosis of patients with lung adenocarcinoma. USP4 can be used as a potential therapeutic target for clinical diagnosis of lung adenocarcinoma and provide a new opportunity for clinical research on lung adenocarcinoma.
Collapse
Affiliation(s)
- Yamin Wei
- Department of Pathology, Affiliated Hospital, Guilin Medical University, Guilin, 541001, China
| | - Shanwang Wei
- Clinical School of Medicine, Qinghai University, Xining, 810000, China
| | - Zhongteng Lei
- Department of Information, Guilin People's Hospital, Guilin, 541001, China
| | - Yan Zhang
- Department of Pathology, Affiliated Hospital, Guilin Medical University, Guilin, 541001, China
| | - Jinxiao Wu
- Department of Pathology, Affiliated Hospital, Guilin Medical University, Guilin, 541001, China
| | - Jinli Huang
- Department of Pathology, Affiliated Hospital, Guilin Medical University, Guilin, 541001, China
| | - Lijuan Fu
- Department of Pathology, Affiliated Hospital, Guilin Medical University, Guilin, 541001, China
| | - Zhimeng Li
- Department of Pathology, Affiliated Hospital, Guilin Medical University, Guilin, 541001, China
| | - Guiying Huang
- Department of Pathology, Affiliated Hospital, Guilin Medical University, Guilin, 541001, China
| | - Yuanna Liang
- Department of Pathology, Affiliated Hospital, Guilin Medical University, Guilin, 541001, China
| | - Jinhua Zheng
- Department of Pathology, Affiliated Hospital, Guilin Medical University, Guilin, 541001, China.
| |
Collapse
|
9
|
Guo R, Shi L, Chen Y, Lin C, Yin W. Exploring the roles of ncRNAs in prostate cancer via the PI3K/AKT/mTOR signaling pathway. Front Immunol 2025; 16:1525741. [PMID: 40170845 PMCID: PMC11959002 DOI: 10.3389/fimmu.2025.1525741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 02/27/2025] [Indexed: 04/03/2025] Open
Abstract
Although various treatment options are available for prostate cancer (PCa), including androgen deprivation therapy (ADT) and chemotherapy, these approaches have not achieved the desired results clinically, especially in the treatment of advanced chemotherapy-resistant PCa. The PI3K/AKT/mTOR (PAM) signaling pathway is a classical pathway that is aberrantly activated in cancer cells and promotes the tumorigenesis, metastasis, resistance to castration therapy, chemoresistance, and recurrence of PCa. Noncoding RNAs (ncRNAs) are a class of RNAs that do not encode proteins. However, some ncRNAs have recently been shown to be differentially expressed in tumor tissues compared with noncancerous tissues and play important roles at the transcription and posttranscription levels. Among the types of ncRNAs, long noncoding RNAs (lncRNAs), microRNAs (miRNAs), circular RNAs (circRNAs), and Piwi-interacting RNAs (piRNAs) can participate in the PAM pathway to regulate PCa growth, metastasis, angiogenesis, and tumor stemness. Therefore, ncRNA therapy that targets the PAM signaling pathway is expected to be a novel and effective approach for treating PCa. In this paper, we summarize the types of ncRNAs that are associated with the PAM pathway in PCa cells as well as the functions and clinical roles of these ncRNAs in PCa. We hope to provide novel and effective strategies for the clinical diagnosis and treatment of PCa.
Collapse
Affiliation(s)
- Rongwang Guo
- Nanchang University, 999 University Avenue, Honggutan District, Nanchang, China
| | - Liji Shi
- School of Chemical and Biological Engineering, Yichun College, Yichun, China
| | - Yonghui Chen
- School of Chemical and Biological Engineering, Yichun College, Yichun, China
| | - Canling Lin
- School of Chemical and Biological Engineering, Yichun College, Yichun, China
| | - Weihua Yin
- Department of Oncology, Baoan Central Hospital of Shenzhen, Bao’ an District, Shenzhen, China
| |
Collapse
|
10
|
Yang X, Wei H, Li J, Li G, Zhang Y, Li H. Efficacy of sialic acid supplementation in early life in autism model rats. Sci Rep 2025; 15:8576. [PMID: 40075137 PMCID: PMC11903695 DOI: 10.1038/s41598-025-93550-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/07/2025] [Indexed: 03/14/2025] Open
Abstract
Autism spectrum disorder (ASD) is a set of heterogeneous neurodevelopmental conditions, the etiology of which remains elusive. Sialic acid (SA) is an essential nutrient for nervous system development, and previous studies reported that the levels of SA were decreased in the blood and saliva of ASD children. However, it is not clear whether SA supplementation can alleviate behavioral problems in autism. We administered SA intervention in the VPA-induced autism model rats, evaluated behavior performance, and measured the levels of Gne and St8sia2 genes, BDNF and anti-GM1. At the same time, untargeted metabolomics was used to characterize the metabolites. It was found that the stereotypical behaviors, social preference and cognitive function were improved after SA supplementation. Additionally, the number of hippocampal neurons was increased, and the shape was normalized. Moreover, 94 differentially abundant metabolites were identified between the high dose SA and VPA groups. These changes in metabolites were correlated with pyrimidine metabolism, lysine degradation metabolism, biosynthesis of amino acids, mineral absorption, protein digestion and absorption, galactose metabolism, phenylalanine, tyrosine and tryptophan biosynthesis and phenylalanine metabolism. In conclusion, SA could ameliorate ASD-like phenotypes and change metabolites in autistic animals, which suggests that it may be a therapeutic approach for ASD.
Collapse
Affiliation(s)
- Xiaolei Yang
- Department of Preventive Medicine, School of Public Health, Qiqihar Medical University, Qiqihar, 161006, China
| | - Hongjuan Wei
- Nursing School, Qiqihar Medical University, Qiqihar, 161006, China
| | - Jiyuan Li
- Department of Preventive Medicine, School of Public Health, Qiqihar Medical University, Qiqihar, 161006, China
| | - Gang Li
- Department of Preventive Medicine, School of Public Health, Qiqihar Medical University, Qiqihar, 161006, China
| | - Yan Zhang
- Nursing School, Qiqihar Medical University, Qiqihar, 161006, China
| | - Hongjie Li
- Department of Preventive Medicine, School of Public Health, Qiqihar Medical University, Qiqihar, 161006, China.
| |
Collapse
|
11
|
Gerlza T, Trojacher C, Fuchs T, Atlic A, Weis R, Adage T, Kungl AJ. Designing a CXCL8-hsa chimera as potential immunmodulator of the tumor micro-environment. Front Immunol 2025; 16:1539733. [PMID: 40124384 PMCID: PMC11926544 DOI: 10.3389/fimmu.2025.1539733] [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: 12/04/2024] [Accepted: 02/13/2025] [Indexed: 03/25/2025] Open
Abstract
Introduction CXCL8, belonging to inflammatory chemokines, is expressed by various cell types and plays a key role in leukocyte trafficking during infections, inflammatory processes, tissue injury and tumor progression. Chemokines interact not only with G-protein coupled receptors but also with glycosaminoglycans (GAGs), which are polyanionic linear polysaccharides. Chemokine-GAG interactions are critical for creating localized concentration gradients, protecting chemokines from degradation, and maintaining their efficacy in vivo. Methods We have previously engineered a CXCL8-based dominant-negative decoy ("PA401") with strongly increased GAG binding affinity combined with complete GPCR knockout, which was originally developed for the treatment of COPD. Here we have optimized our engineering protocol by minimizing CXCL8 mutations while conserving its in vitro dominant-negative activities. This novel CXCL8-based decoy (mtCXCL8) was further fused to human serum albumin (HSA) to overcome the typically very short serum half-life of chemokine-based biologics. We are therefore able to present here an entirely novel CXCL8-based biologic (hsa/mtCXCL8) which reflects our threefold modification strategy - increasing GAG-binding affinity by minimal mutagenesis, GPCR knockout, and fusion to HSA - thus representing a comprehensive and novel approach towards addressing chronic CXCL8-driven diseases. Results In the current study, we have investigated the immunomodulatory potential of our new decoy in a 3-D cellular tumor model ("BioMAP") which relates the biomarker interaction profile of immune and tumor cells to a data-base mirrored biomarker read-out. The obtained BioMAP results suggest an impact of hsa/mtCXCL8 on the immune compartment of the VascHT29 cell model by modulating cytokine levels and inhibiting immune cell activation markers. When combined with Keytruda (Pembrolizumab), a PD-1 inhibitor, it enhances some of its known activities, indicating potential synergistic effects, but further investigation is needed due to the observed increase in soluble IL-6 and limitations in dose selection for future in vivo studies. Discussion By prolonging the presence of engineered chemokine mutants in the bloodstream and optimizing their stability, these strategies aim to enhance the therapeutic efficacy of CXCL8-based interventions, offering promising avenues for the treatment of several CXCL8-mediated pathologies, including cancer.
Collapse
Affiliation(s)
- Tanja Gerlza
- Karl-Franzens-University Graz, Institute of Pharmaceutical Sciences, Pharmaceutical Chemistry, Graz, Austria
| | - Christina Trojacher
- Karl-Franzens-University Graz, Institute of Pharmaceutical Sciences, Pharmaceutical Chemistry, Graz, Austria
| | - Thomas Fuchs
- Medical University Graz, Otto Loewi Research Center, Graz, Austria
| | | | | | | | - Andreas J. Kungl
- Karl-Franzens-University Graz, Institute of Pharmaceutical Sciences, Pharmaceutical Chemistry, Graz, Austria
- Antagonis Biotherapeutics GmbH, Graz, Austria
| |
Collapse
|
12
|
Liu X, Zhang X, Zeng T, Chen Y, Ye L, Wang S, Li Y. FOSL1 drives the malignant progression of pancreatic cancer cells by regulating cell stemness, metastasis and multidrug efflux system. J Transl Med 2025; 23:268. [PMID: 40038751 PMCID: PMC11877717 DOI: 10.1186/s12967-025-06304-w] [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: 09/24/2024] [Accepted: 02/23/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Targeted therapy is an effective strategy for the treatment of advanced and metastatic pancreatic cancer, one of the leading causes for cancer-related death worldwide. To address the limitations of existing targeted drugs, there is an urgently need to find novel targets and therapeutic strategies. Transcription factor FOS like 1 (FOSL1) is a potential therapeutic target for challenging pancreatic cancer, which contributes to the malignant progression and poor gnosis of pancreatic cancer. High mobility group A1 (HMGA1) is a nonhistone chromatin structural protein that contributes to malignant progression and poor prognosis of cancer. METHODS Human FOSL1 complete RNA, shRNA against FOSL1 and shRNA against HMGA1 lentiviral recombination vectors were used to overexpress FOSL1 and knock down FOSL1 and HMGA1. RNA sequencing, Q-PCR and Western blots were used to investigate the mechanism of FOSL1 in regulating the proliferation of pancreatic cancer cells. The relationship between FOSL1 and HMGA1 were analyzed by co-immunoprecipitation Mass spectrometry, Q-PCR of chromatin immunoprecipitation and Western blots. The regulation of FOSL1 and HMGA1 in the invasion and migration, stemness, and multidrug efflux system were determined by transwell assay, sphere formation assay, immunofluorescence, Q-PCR and Western blots. RESULTS We found that FOSL1 promoted the proliferation and progression of pancreatic cancer by trigging stemness, invasion and metastasis, and drug resistance. HMGA1 was a key downstream target regulated by FOSL1 at the transcriptional level and directly interacted with FOSL1. Knockdown of HMGA1 inhibited the proliferation of pancreatic cancer cells by regulating the expression of genes related to stemness, epithelial-mesenchymal transition and multidrug efflux system. Targeted inhibition of FOSL1 and HMGA1 expression significantly inhibited the proliferation of pancreatic cancer cells. CONCLUSION FOSL1 promote the malignant progression of pancreatic cancer by promoting HMGA1 expression. Targeting FOSL1 and HMGA1 in monotherapy or combination therapy is a promising strategy for the treatment of advanced and metastasis pancreatic cancer.
Collapse
Affiliation(s)
- Xiaolong Liu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China
| | - Xueyan Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Tingyu Zeng
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Yali Chen
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Liu Ye
- Medical College of Guizhou University, Guiyang, 550025, Guizhou Province, China
| | - Shuping Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China.
| | - Yulan Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China.
| |
Collapse
|
13
|
Bai L, Qian X, Zhang H, Yuan Y, Cui X, Cheng M, Han Y. Plectin, a novel regulator in migration, invasion and adhesion of ovarian cancer. Cell Biosci 2025; 15:15. [PMID: 39915800 PMCID: PMC11804098 DOI: 10.1186/s13578-025-01349-2] [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: 04/08/2024] [Accepted: 01/11/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Ovarian cancer (OC) is one of the most prevalent gynecologic malignancies and exhibites the highest fatality rate among all gynecologic malignancies. The absence of an early diagnostic biomarker and therapeutic target contributes to an overall 5-year survival rate ranging from 30 to 50%. Plectin (PLEC), a 500 kDa scaffolding protein, has gained prominence in recent years due to its pivotal role in various cellular biological functions such as cell morphology, migration and adhesion, while the accurate role of PLEC in OC remains elusive. RESULTS In this study, our findings demonstrate that PLEC exerts a positive influence on the progression of OC, encompassing cellular proliferation, migration, invasion, and adhesion both in vitro and in vivo. CONCLUSIONS The results providing new insights for the diagnosis and treatment in OC.
Collapse
Affiliation(s)
- Lanning Bai
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, Shandong, 261053, P. R. China
| | - Xueqian Qian
- School of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P. R. China
| | - Hui Zhang
- School of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P. R. China
| | - Yi Yuan
- School of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P. R. China
| | - Xiaodong Cui
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, Shandong, 261053, P. R. China.
| | - Min Cheng
- Department of Physiology, Shandong Second Medical University, Weifang, Shandong, 261053, P. R. China.
| | - Yangyang Han
- School of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, 261053, P. R. China.
| |
Collapse
|
14
|
Torre-Cea I, Berlana-Galán P, Guerra-Paes E, Cáceres-Calle D, Carrera-Aguado I, Marcos-Zazo L, Sánchez-Juanes F, Muñoz-Félix JM. Basement membranes in lung metastasis growth and progression. Matrix Biol 2025; 135:135-152. [PMID: 39719224 DOI: 10.1016/j.matbio.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 12/26/2024]
Abstract
The lung is a highly vascularized tissue that often harbors metastases from various extrathoracic malignancies. Lung parenchyma consists of a complex network of alveolar epithelial cells and microvessels, structured within an architecture defined by basement membranes. Consequently, understanding the role of the extracellular matrix (ECM) in the growth of lung metastases is essential to uncover the biology of this pathology and developing targeted therapies. These basement membranes play a critical role in the progression of lung metastases, influencing multiple stages of the metastatic cascade, from the acquisition of an aggressive phenotype to intravasation, extravasation and colonization of secondary sites. This review examines the biological composition of basement membranes, focusing on their core components-collagens, fibronectin, and laminin-and their specific roles in cancer progression. Additionally, we discuss the function of integrins as primary mediators of cell adhesion and signaling between tumor cells, basement membranes and the extracellular matrix, as well as their implications for metastatic growth in the lung. We also explore vascular co-option (VCO) as a form of tumor growth resistance linked to basement membranes and tumor vasculature. Finally, the review covers current clinical therapies targeting tumor adhesion, extracellular matrix remodeling, and vascular development, aiming to improve the precision and effectiveness of treatments against lung metastases.
Collapse
Affiliation(s)
- Irene Torre-Cea
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain
| | - Patricia Berlana-Galán
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain
| | - Elena Guerra-Paes
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain
| | - Daniel Cáceres-Calle
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain
| | - Iván Carrera-Aguado
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain
| | - Laura Marcos-Zazo
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain
| | - Fernando Sánchez-Juanes
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain.
| | - José M Muñoz-Félix
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain.
| |
Collapse
|
15
|
Cáceres-Calle D, Torre-Cea I, Marcos-Zazo L, Carrera-Aguado I, Guerra-Paes E, Berlana-Galán P, Muñoz-Félix JM, Sánchez-Juanes F. Integrins as Key Mediators of Metastasis. Int J Mol Sci 2025; 26:904. [PMID: 39940673 PMCID: PMC11816423 DOI: 10.3390/ijms26030904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/16/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Metastasis is currently becoming a major clinical concern, due to its potential to cause therapeutic resistance. Its development involves a series of phases that describe the metastatic cascade: preparation of the pre-metastatic niche, epithelial-mesenchymal transition, dissemination, latency and colonization of the new tissue. In the last few years, new therapeutic targets, such as integrins, are arising to face this disease. Integrins are transmembrane proteins found in every cell that have a key role in the metastatic cascade. They intervene in adhesion and intracellular signaling dependent on the extracellular matrix and cytokines found in the microenvironment. In this case, integrins can initiate the epithelial-mesenchymal transition, guide the formation of the pre-metastatic niche and increase tumor migration and survival. Integrins also take part in the tumor vascularization process necessary to sustain metastasis. This fact emphasizes the importance of inhibitory therapies capable of interfering with the function of integrins in metastasis.
Collapse
Affiliation(s)
- Daniel Cáceres-Calle
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (D.C.-C.); (I.T.-C.); (L.M.-Z.); (I.C.-A.); (E.G.-P.); (P.B.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Irene Torre-Cea
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (D.C.-C.); (I.T.-C.); (L.M.-Z.); (I.C.-A.); (E.G.-P.); (P.B.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Laura Marcos-Zazo
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (D.C.-C.); (I.T.-C.); (L.M.-Z.); (I.C.-A.); (E.G.-P.); (P.B.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Iván Carrera-Aguado
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (D.C.-C.); (I.T.-C.); (L.M.-Z.); (I.C.-A.); (E.G.-P.); (P.B.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Elena Guerra-Paes
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (D.C.-C.); (I.T.-C.); (L.M.-Z.); (I.C.-A.); (E.G.-P.); (P.B.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Patricia Berlana-Galán
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (D.C.-C.); (I.T.-C.); (L.M.-Z.); (I.C.-A.); (E.G.-P.); (P.B.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - José M. Muñoz-Félix
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (D.C.-C.); (I.T.-C.); (L.M.-Z.); (I.C.-A.); (E.G.-P.); (P.B.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Fernando Sánchez-Juanes
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (D.C.-C.); (I.T.-C.); (L.M.-Z.); (I.C.-A.); (E.G.-P.); (P.B.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| |
Collapse
|
16
|
Wu L, Liu C, Hu W. Comprehensive investigation of matrix metalloproteinases in skin cutaneous melanoma: diagnostic, prognostic, and therapeutic insights. Sci Rep 2025; 15:2152. [PMID: 39820824 PMCID: PMC11739484 DOI: 10.1038/s41598-025-85887-2] [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: 10/08/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025] Open
Abstract
The dysregulation of matrix metalloproteinases (MMPs) in skin cutaneous melanoma (SKCM) represents a critical aspect of tumorigenesis. In this study, we investigated the diagnostic, prognostic, and therapeutic aspects of the MMPs in SKCM. Thirteen SKCM cell lines and seven normal skin cell lines were cultured under standard conditions for experimental analyses. RNA and DNA were extracted, followed by RT-qPCR to assess MMP expression and promoter methylation analysis to determine methylation levels. Functional assays, including cell proliferation, colony formation, and wound healing, were conducted post-MMP7 knockdown using siRNA in A375 cells. Databases like GEPIA2, HPA, MEXPRESS, and miRNet were employed for expression, survival, methylation, and miRNA-mRNA network analyses. We investigated the expression and promoter methylation landscape of MMPs in SKCM cell lines, revealing significant (p-value < 0.05) up-regulation of MMP1, MMP7, MMP9, MMP10, MMP11, MMP12, MMP13, MMP14, and MMP25, alongside down-regulation of MMP2, MMP3, and MMP21. Furthermore, our analysis demonstrated a significant (p-value < 0.05) inverse correlation between MMP expression levels and promoter methylation status, suggesting a potential regulatory role of DNA methylation in MMP dysregulation. Notably, MMP7, MMP11, and MMP14 exhibited significant (p-value < 0.05) associations with the overall survival of SKCM patients, emphasizing their prognostic significance. Additionally, Receiver operating characteristic (ROC) curve analysis highlighted the significant (p-value < 0.05) diagnostic potential of MMP7, MMP11, and MMP14 in distinguishing SKCM from normal individuals. Subsequent validation across multiple cohorts confirmed significant (p-value < 0.05) elevated MMP expression levels in SKCM tissues, particularly in advanced disease stages, further emphasizing their role in tumor progression. Furthermore, we elucidated potential regulatory pathways involving miR-22-3p, which targets MMP7, MMP11, and MMP14 genes in SKCM. Our findings also revealed associations between MMP expression and immune modulation, drug sensitivity, and functional states of SKCM cells. Lastly, MMP7 knockdown in A375 cells significantly significant (p-value < 0.05) impacted several characteristics, including cell proliferation, colony formation, and wound healing. Our findings highlight the diagnostic, prognostic, and therapeutic potential of MMP7, MMP11, and MMP14 in SKCM. These MMPs could serve as biomarkers for early detection and targets for therapy. Future efforts should focus on preclinical and clinical validation to translate these insights into personalized diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Lingxia Wu
- Dermatology, Changzhi Second People's Hospital, Changzhi, 046000, Shanxi, China
| | - Chenxiaoxiao Liu
- The First Clinical Institute, Zunyi Medical University, Zunyi, 520300, Guizhou, China
| | - Weicai Hu
- Dermatology, Changzhi Second People's Hospital, Changzhi, 046000, Shanxi, China.
| |
Collapse
|
17
|
Alshahrani MY, Oghenemaro EF, Rizaev J, Kyada A, Roopashree R, Kumar S, Taha ZA, Yadav G, Mustafa YF, Abosaoda MK. Exploring the modulation of TLR4 and its associated ncRNAs in cancer immunopathogenesis, with an emphasis on the therapeutic implications and mechanisms underlying drug resistance. Hum Immunol 2025; 86:111188. [PMID: 39631102 DOI: 10.1016/j.humimm.2024.111188] [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] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024]
Abstract
This study provides an in-depth analysis of the pathogenic relevance, therapeutic implications, and mechanisms of treatment resistance associated with TLR4 and its ncRNAs in cancer immunopathogenesis. TLR4, a pivotal component of the innate immune response, has been implicated in promoting inflammation, tumorigenesis, and immune evasion across various malignancies, including gastric, ovarian, and hepatocellular carcinoma. The interactions between TLR4 and specific ncRNAs, such as lncRNAs and miRNAs, play a crucial role in modulating TLR4 signaling pathways, influencing immune cell dynamics, and contributing to chemoresistance. These ncRNAs facilitate tumor-promoting processes, including macrophage polarization, dendritic cell suppression, and T-cell regulation, effectively establishing an immunosuppressive tumor microenvironment that further enhances therapeutic resistance. A comprehensive understanding of the complex interplay between TLR4 and ncRNAs unveils potential avenues for identifying predictive biomarkers and discovering novel therapeutic targets in cancer. Future research initiatives should prioritize the development of personalized therapeutic strategies that specifically target TLR4 signaling and its ncRNA regulators to counteract drug resistance and improve clinical outcomes. This review extensively evaluates the role of TLR4 in cancer biology, emphasizing its critical importance in developing innovative cancer management strategies.
Collapse
Affiliation(s)
- Mohammad Y Alshahrani
- Central Labs, King Khalid University, AlQura 'a, Abha, P.O. Box 960, Saudi Arabia; Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Enwa Felix Oghenemaro
- Delta State University, Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Abraka, Delta State, Nigeria.
| | - Jasur Rizaev
- Professor, Doctor of Medical Sciences, Department of Public Health and Healthcare Management, Rector, Samarkand State Medical University, 18, Amir Temur Street, Samarkand, Uzbekistan.
| | - Ashishkumar Kyada
- Marwadi University, Research Center, Department of Pharmacy, Faculty of Health Sciences, Marwadi University, Rajkot 360003, Gujarat, India.
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India.
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Zahraa Ahmed Taha
- Medical Laboratory Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, 51001 Babylon, Iraq.
| | - Geeta Yadav
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India.
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, -41001, Iraq.
| | - Munthar Kadhim Abosaoda
- College of Pharmacy, The Islamic University, Najaf, Iraq; College of Pharmacy, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; College of Pharmacy, The Islamic University of Babylon, Babylon, Iraq.
| |
Collapse
|
18
|
Lu P, Yang L, Chen W, Li K, Chen X, Qu S. Four-dimensional trapped ion mobility spectrometry proteomics reveals circulating extracellular vesicles encapsulated drivers of nasopharyngeal carcinoma distant dissemination. Talanta 2025; 282:126907. [PMID: 39341061 DOI: 10.1016/j.talanta.2024.126907] [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: 08/17/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024]
Abstract
Nasopharyngeal carcinoma (NPC) is a head and neck cancer with a high propensity for early metastatic spread. Emerging evidence shows that extracellular vesicles (EVs) are key players in cancer metastasis, but their role in NPC metastasis remains poorly understood. We here present the first description of the proteomic and functional profiles of serum-derived circulating small EVs in metastatic NPC patients. To enhance the capture of low-abundance signaling proteins in EVs, timsTOF-based four-dimensional label-free quantitative proteomics was employed. We found that metastatic NPC patients (M-NPC-EVs) exhibited the highest serum EV levels compared to locoregional patients (L-NPC-EVs) and healthy subjects (Normal-EVs). The proteome of M-NPC-EVs differed substantially from L-NPC-EVs and was functionally enriched in pathways regulating cell polarity and motility, glucose metabolism, and angiogenesis. Functional assays testing individual EV samples demonstrated that M-NPC-EVs pronouncedly enhanced NPC cell migration, invasion, and the formation of lamellipodia and filopodia in vitro, and promoted angiogenesis in subcutaneous Matrigel plugs in vivo. In silico analyses suggested that PTPRA, TPI1 and GPI highly enriched in M-NPC-EVs were putative drivers underlying the motogenic and angiogenic activities of M-NPC-EVs, and their high expression levels were associated with a poor prognosis of NPC patients. The increased expression of PTPRA, TPI1 and GPI in M-NPC-EVs was then validated in an independent cohort consisting of 175 NPC patients (locoregional n = 114; metastatic n = 61). Together, utilizing patient-derived EVs, we mimicked the potential pro-metastatic functions of EVs in NPC patients in vitro and in vivo and provided novel insights into their bioactive cargoes.
Collapse
Affiliation(s)
- Pingan Lu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, 530021, Guangxi Autonomous Region, China
| | - Liu Yang
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, 530021, Guangxi Autonomous Region, China
| | - Weiling Chen
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, 530021, Guangxi Autonomous Region, China
| | - Kaiguo Li
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, 530021, Guangxi Autonomous Region, China
| | - Xuxia Chen
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, 530021, Guangxi Autonomous Region, China
| | - Song Qu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, 530021, Guangxi Autonomous Region, China; Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, China; Guangxi Key Laboratory of High-Incidence Tumor Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, China; Guangxi Nasopharyngeal Carcinoma Clinical Research Center, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
19
|
Zhang S, Xie R, Wang L, Fu G, Zhang C, Zhang Y, Yu J. TMEM252 inhibits epithelial-mesenchymal transition and progression in papillary thyroid carcinoma by regulating Notch1 expression. Head Neck 2025; 47:324-338. [PMID: 39152570 DOI: 10.1002/hed.27922] [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: 03/28/2024] [Revised: 07/23/2024] [Accepted: 08/07/2024] [Indexed: 08/19/2024] Open
Abstract
BACKGROUND Papillary thyroid carcinoma (PTC) accounts for about 85% of thyroid cancer cases. Transmembrane protein 252 (TMEM252) is a gene encoding a transmembrane protein that has only been reported to be associated with triple-negative breast cancer. Herein, we first elucidated the physiological roles and possible regulatory proteins of TMEM252 in PTC pathogenesis. METHODS Quantitative real-time polymerase chain reaction, western blot, and immunohistochemical analyses were utilized to ascertain the relative TMEM252 expression in PTC and surrounding normal tissues. Functional investigations involved CCK-8 viability assay, EdU incorporation assay for proliferation, transwell assays for migration and invasion, and an in vivo tumor development assessment to evaluate the TMEM252-mediated regulation of tumor formation. RESULTS Our results first revealed diminished TMEM252 transcript and protein expressions in PTC tissues and cell lines. TMEM252 overexpression suppressed cell proliferation through reducing p53, p21, and p16 expression. Conversely, TMEM252 depletion has opposite effects in PTC cells both in vivo. Additionally, the upregulation of TMEM252 demonstrated cell migration and invasion suppression by impeding the epithelial-mesenchymal transition (EMT) process via inhibition of the Notch pathway. Furthermore, overexpression of TMEM252 suppressed tumor growth in vivo. CONCLUSION Our study elucidates that TMEM252 suppresses PTC progression by modulating the Notch pathway. These findings underscore TMEM252 is a potential therapeutic target in managing PTC.
Collapse
Affiliation(s)
- Shuyong Zhang
- Department of Thyroid Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Rong Xie
- Department of Thyroid Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Liuhuan Wang
- Department of Thyroid Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Guoxue Fu
- Department of Thyroid Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chenxi Zhang
- Department of Thyroid Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yang Zhang
- Department of Thyroid Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jichun Yu
- Department of Thyroid Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
20
|
Martínez-López N, Pereiro P, Saco A, Lama R, Figueras A, Novoa B. Characterization of a fish-specific immunoglobulin-like domain-containing protein (Igldcp) in zebrafish (Danio rerio) induced after nodavirus infection. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 162:105285. [PMID: 39515405 DOI: 10.1016/j.dci.2024.105285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 11/05/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
One of the most highly induced genes in zebrafish (Danio rerio) larvae after infection with the nodavirus red-spotted grouper nervous necrosis virus (RGNNV) was a member of the immunoglobulin superfamily (IgSF), which has remained uncharacterized and erroneously annotated in zebrafish and other fish species as galectin 17 (lgals17). We characterized this gene and named it immunoglobulin (Ig)-like domain-containing protein (igldcp), a new member of the IgSF that does not possess orthologs in mammals. Igldcp expression is induced by viral infection and it belongs to the group of interferon-stimulated genes (ISGs). In vitro overexpression of igldcp decreased RGNNV replication, whereas in vivo knockdown of this gene had the opposite effect, resulting in increased larval mortality. RNA-Seq analyses of larvae overexpressing igldcp in the absence or presence of infection with RGNNV showed that the main processes affected by Igldcp could be directly involved in the regulation of various cellular processes associated with the modulation of the immune system.
Collapse
Affiliation(s)
| | | | - Amaro Saco
- Institute of Marine Research (IIM-CSIC), Vigo, Spain
| | - Raquel Lama
- Institute of Marine Research (IIM-CSIC), Vigo, Spain
| | | | - Beatriz Novoa
- Institute of Marine Research (IIM-CSIC), Vigo, Spain.
| |
Collapse
|
21
|
Ávalos-Navarro G, Bautista-Herrera LA, Garibaldi-Ríos AF, Ramírez-Patiño R, Gutiérrez-García M, Briseño-Álvarez P, Jave-Suárez LF, Reyes-Uribe E, Gallegos-Arreola MP. Serum α1-AT Levels and SERPINA1 Molecular Analysis in Breast Cancer: An Experimental and Computational Study. Diseases 2024; 13:1. [PMID: 39851465 PMCID: PMC11765096 DOI: 10.3390/diseases13010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/11/2024] [Accepted: 12/17/2024] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND/OBJECTIVES Breast cancer (BC) is a heterogeneous disease with multifactorial origins, including environmental, genetic, and immunological factors. Inflammatory cytokines, such as alpha 1 antitrypsin (α1-AT), are increased in BC and affect physiological and pathological conditions. This study aimed to evaluate the serum levels of α1-AT and perform a computational analysis of SERPINA1 in BC, as well as their association with molecular subtypes and clinical features. METHODS For the experimental analysis, we evaluated 255 women with BC and 53 healthy women (HW) in a cross-sectional study. Molecular subtypes were identified by immunohistochemistry and TNM was used for clinical staging. Soluble levels of α1-AT were quantified by ELISA. Computational analysis of SERPINA1 expression was performed using GEPIA and cBioPortal. RESULTS α1-AT was increased in BC women versus HW (75.8 ng/mL vs. 532.2 ng/mL). Luminal A had higher concentration (547.5 ng/mL) than Triple Negative (TN) (484.1 ng/mL), but the levels were not associated with clinical stage. The computational analysis showed that SERPINA1 is overexpressed in BC with differential expression among subtypes; its overexpression is associated with a better prognosis, longer disease-free survival, and overall survival. CONCLUSIONS α1-AT levels are increased in women with BC women compared to HW. The Luminal A subtype shows higher soluble protein levels than the TN one. Furthermore, SERPINA1 mRNA overexpression in BC is linked to a protective effect.
Collapse
Affiliation(s)
- Guadalupe Ávalos-Navarro
- Departamento de Ciencias Médicas y de la Vida, Centro Universitario de la Ciénega (CUCIÉNEGA), Universidad de Guadalajara, Av. Universidad 1115, Lindavista, Ocotlán 47820, Jalisco, Mexico; (G.Á.-N.); (R.R.-P.); (E.R.-U.)
| | - Luis A. Bautista-Herrera
- Departamento de Farmacobiología, Centro Universitario de Ciencias Exactas e Ingenierías (CUCEI), Universidad de Guadalajara, Blvd. Marcelino García Barragán 1421, Olímpica, Guadalajara 44430, Jalisco, Mexico;
| | - Asbiel Felipe Garibaldi-Ríos
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdeG), Guadalajara 44340, Jalisco, Mexico;
- División de Genética, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Sierra Mojada 800, Independencia Oriente, Guadalajara 44340, Jalisco, Mexico
| | - Ramiro Ramírez-Patiño
- Departamento de Ciencias Médicas y de la Vida, Centro Universitario de la Ciénega (CUCIÉNEGA), Universidad de Guadalajara, Av. Universidad 1115, Lindavista, Ocotlán 47820, Jalisco, Mexico; (G.Á.-N.); (R.R.-P.); (E.R.-U.)
| | - Marisol Gutiérrez-García
- Licenciatura en Químico Farmacéutico Biólogo, Centro Universitario de Ciencias Exactas e Ingenierías (CUCEI), Universidad de Guadalajara, Blvd. Marcelino García Barragán 1421, Olímpica, Guadalajara 44430, Jalisco, Mexico;
| | - Perla Briseño-Álvarez
- Licenciatura en Químico Farmacéutico Biólogo, Centro Universitario de la Ciénega (CUCIÉNEGA), Universidad de Guadalajara, Av. Universidad 1115, Lindavista, Ocotlán 47820, Jalisco, Mexico;
| | - Luis Felipe Jave-Suárez
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara 44340, Jalisco, Mexico;
| | - Emmanuel Reyes-Uribe
- Departamento de Ciencias Médicas y de la Vida, Centro Universitario de la Ciénega (CUCIÉNEGA), Universidad de Guadalajara, Av. Universidad 1115, Lindavista, Ocotlán 47820, Jalisco, Mexico; (G.Á.-N.); (R.R.-P.); (E.R.-U.)
| | - Martha Patricia Gallegos-Arreola
- División de Genética, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Sierra Mojada 800, Independencia Oriente, Guadalajara 44340, Jalisco, Mexico
| |
Collapse
|
22
|
Ni X, Pan F, Lang YK, Zhang W. Prognostic significance of NUAK1 and its association with immune infiltration in stomach adenocarcinoma. Discov Oncol 2024; 15:800. [PMID: 39692916 DOI: 10.1007/s12672-024-01688-5] [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: 10/05/2024] [Accepted: 12/09/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Stomach adenocarcinoma (STAD) represents a significant global health burden, accounting for a considerable proportion of cancer-related mortalities, and NUAK1, a protein kinase, plays a crucial role in cellular metabolism, cell cycle regulation, migration, and tumor progression. However, its relationship with prognosis and immune infiltration in STAD has not been thoroughly investigated. METHODS RNA sequencing data from the Cancer Genome Atlas (TCGA) and Genotypic Tissue Expression Project (GTEx) databases were employed to assess disparities in NUAK1 expression between STAD tumour and normal tissues. Additionally, we investigated the correlation between NUAK1 expression and patient prognosis, in addition to the level of immune cell infiltration. The potential functions were elucidated through an examination of the Gene Ontology (GO) Encyclopedia, the Kyoto Encyclopedia of Genes and Genomes (KEGG), and an enrichment analysis (GSEA). The GeneMANIA was used to validate the functions of nuak1-related genes. RESULTS Our analysis demonstrated that NUAK1 expression in tumour tissues exhibited a notable disparity from that observed in normal tissues, with elevated levels detected in STAD tissues. We used the GeneMANIA database to identify functionally similar genes with significantly higher expression for some genes in the unpaired group samples. An elevated NUAK1 expression level was found to correlate with a poorer overall survival (OS), disease-specific survival (DSS), and progression-free intervals (PFI). Additionally, immune infiltration analysis indicated a significant positive correlation between NUAK1 expression and various tumor-infiltrating immune cells, while a negative correlation was observed with T helper cell 17(Th17) cells. Furthermore, enrichment analysis was conducted to identify relevant biological features and pathways. CONCLUSION The expression levels of NUAK1 are significantly increased in STAD, and this heightened expression correlates with diminished OS, DSS, and PFI among affected patients. These observations indicate that NUAK1 has the potential to function as a prognostic biomarker for STAD and may represent a viable therapeutic target for intervention in its management.
Collapse
Affiliation(s)
- Xin Ni
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Zhenjiang, Jiangsu, China
| | - Fan Pan
- Department of Articular Surgery, Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Zhenjiang, Jiangsu, China
| | | | - Wei Zhang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Zhenjiang, Jiangsu, China.
| |
Collapse
|
23
|
Liu Z, Yang L, Ni Y, Chen K, Yan Q, Zhao Z, Xu B, Li Y, Li R, Li J. Enhanced bacteriostasis and osseointegrative properties of SiRNA-modified polyetheretherketone surface for implant applications. PLoS One 2024; 19:e0314091. [PMID: 39636795 PMCID: PMC11620434 DOI: 10.1371/journal.pone.0314091] [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: 06/26/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024] Open
Abstract
Polyetheretherketone (PEEK), bearing an elastic modulus that effectively simulates the innate properties of natural bone, has come into the spotlight as a promising bone substitute material. Nonetheless, the biologically inert nature of PEEK, combined with its insubstantial osseointegration and sterilization capabilities, pose constraints on its clinical application in the realm of implants. RNA interference (RNAi), an effective technique used for gene expression regulation, has begun to be applied in implant surface modification. Herein, siCKIP-1 is securely affixed to the surface of PEEK implants, aided by an antibacterial polyphenol tannic acid (pTAN) coatings, enhancing physiologic osseointegration and inhibiting bacterial infection. This method breakthrough not merely facilitates the convenience, but also multifaceted PEEK implants' refinements. The modified PEEK implants have impressive biocompatibility coupled with a noteworthy degree of antibacterial properties. Meanwhile, modified PEEK implants improved osteogenic differentiation of rat bone mesenchymal stem cells (rBMSCs) and demonstrated excellent osteointegrative properties in rat femur implantation models. Therefore, identifying a new implant material with excellent biocompatibility and biomechanical properties is essential.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Stomatology, Airforce Medical Center PLA, Air Force Medical University, Beijing, People’s Republic of China
| | - Libin Yang
- Department of Stomatology, Shizuishan Second People’s Hospital, Shizuishan, Ningxia Province, People’s Republic of China
| | - Yazhuo Ni
- Department of prosthodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin, People’s Republic of China
| | - Keying Chen
- Department of prosthodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin, People’s Republic of China
| | - Qiquan Yan
- Department of prosthodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin, People’s Republic of China
| | - Zhiying Zhao
- Baodi Hospital, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Bo Xu
- Department of prosthodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin, People’s Republic of China
| | - Yaoyang Li
- Department of prosthodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin, People’s Republic of China
| | - Rui Li
- Department of prosthodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin, People’s Republic of China
| | - Jianwen Li
- Department of Radiology, Shizuishan Second People’s Hospital, Shizuishan, Ningxia Province, People’s Republic of China
| |
Collapse
|
24
|
Zhang W, Zhang P, Wang X, Lin Y, Xu H, Mao R, Zhu S, Lin T, Cai J, Lin J, Kang M. SORBS2-Mediated inhibition of malignant behaviors in esophageal squamous cell carcinoma through TIMP3. Int Immunopharmacol 2024; 142:113096. [PMID: 39288625 DOI: 10.1016/j.intimp.2024.113096] [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: 06/21/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is characterized by high invasiveness and poor prognosis. The role of Sorbin and SH3 domain-containing protein 2 (SORBS2) in ESCC remains largely unexplored. METHODS The expression levels of SORBS2 in ESCC were detected using RNA-seq and proteomics data. The biological functions of SORBS2 in ESCC were investigated through in vivo and in vitro experiments. The mechanism of SORBS2 was explored using RIP-seq technology, which identified the key downstream molecule metalloproteinase-3 (TIMP3). The interaction between SORBS2 and TIMP3, including specific binding sites, was validated through RIP-qPCR and RNA pull-down assays. The impact of altered SORBS2 expression in ESCC on HUVECs was assessed using endothelial tube formation assays. RESULTS SORBS2 expression was significantly downregulated in ESCC tissues, and its decreased expression was associated with poor prognosis. Overexpression of SORBS2 in ESCC cell lines inhibited cell proliferation, migration, and invasion both in vitro and in vivo. Mechanistically, SORBS2 bound to the 3' UTR of TIMP3 mRNA, enhancing its stability and thereby regulating TIMP3 expression. Rescue experiments demonstrated that increased TIMP3 expression could reverse the promotive effects of SORBS2 knockdown on ESCC, confirming TIMP3 as a critical downstream molecule of SORBS2. Furthermore, downregulation of SORBS2 in ESCC cells was associated with activation of HUVEC functions, whereas upregulation of TIMP3 could reverse this effect. The SORBS2/TIMP3 axis may exert tumor suppressive effects by influencing extracellular matrix degradation. CONCLUSION This study confirms that SORBS2 inhibits ESCC tumor progression by regulating extracellular matrix degradation through TIMP3, providing a potential therapeutic target for future treatment interventions.
Collapse
Affiliation(s)
- Weiguang Zhang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Peipei Zhang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Xiaoqing Wang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ye Lin
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Hui Xu
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Renyan Mao
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Shujing Zhu
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Tianxin Lin
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Junlan Cai
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Jihong Lin
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Clinical Research Center for Thoracic Tumors of Fujian Province, Fuzhou, China.
| | - Mingqiang Kang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of Cardiothoracic Surgery, Affiliated Hospital of Putian University, Putian, China; Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China; Clinical Research Center for Thoracic Tumors of Fujian Province, Fuzhou, China.
| |
Collapse
|
25
|
Li C, Wang X, Shi D, Yang M, Yang W, Chen L. FAM83H regulated by glis3 promotes triple-negative breast cancer tumorigenesis and activates the NF-κB signaling pathway. J Mol Histol 2024; 55:1271-1283. [PMID: 39304594 DOI: 10.1007/s10735-024-10268-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive and invasive form of breast cancer (BC) with a high mortality rate and a lack of effective targeted drugs. Family with sequence similarity 83 member H (FAM83H) is critically implicated in tumorigenesis. However, the potential role of FAM83H in TNBC remains elusive. Here, we discovered that FAM83H exhibited high expression in tumor tissues of patients with TNBC and was associated with TNM stage. Gain- or loss-of-function experiments were conducted to explore the biological role of FAM83H in TNBC. Subsequently, functional enrichment analysis confirmed that FAM83H overexpression promoted TNBC cell proliferation, invasion, migration and epithelial-mesenchymal transition (EMT), accompanied by upregulation of cyclin E, cyclin D, Vimentin, N-cadherin and Slug. As observed, FAM83H knockdown showed anti-cancer effects, such as fostering apoptosis and inhibiting tumorigenicity and metastasis of TNBC cells. Mechanistically, FAM83H activated the NF-κB signaling pathway. Moreover, a dual-luciferase reporter assay demonstrated that GLIS family zinc finger 3 (GLIS3) bound to the promoter of FAM83H and enhanced its transcription. Notably, overexpression of GLIS3 significantly stimulated TNBC cell proliferation and invasion, and all of this was reversed by rescue experiments involving the knockdown of FAM83H. Overall, FAM83H exacerbates tumor progression, and in-depth understanding of FAM83H as a therapeutic target for TNBC will provide clinical translational potential for intervention therapy.
Collapse
Affiliation(s)
- Chenhao Li
- The Second Department of Thyroid and Breast Surgery, Cangzhou Central Hospital, No.16 Xinhua West Road, Cangzhou, Hebei, China
| | - Xin Wang
- The Second Department of Thyroid and Breast Surgery, Cangzhou Central Hospital, No.16 Xinhua West Road, Cangzhou, Hebei, China
| | - Dongliang Shi
- The Second Department of Thyroid and Breast Surgery, Cangzhou Central Hospital, No.16 Xinhua West Road, Cangzhou, Hebei, China
| | - Meng Yang
- The Second Department of Thyroid and Breast Surgery, Cangzhou Central Hospital, No.16 Xinhua West Road, Cangzhou, Hebei, China
| | - Wenhua Yang
- The Second Department of Thyroid and Breast Surgery, Cangzhou Central Hospital, No.16 Xinhua West Road, Cangzhou, Hebei, China
| | - Liang Chen
- The Second Department of Thyroid and Breast Surgery, Cangzhou Central Hospital, No.16 Xinhua West Road, Cangzhou, Hebei, China.
| |
Collapse
|
26
|
Thapa R, Gupta S, Gupta G, Bhat AA, Smriti, Singla M, Ali H, Singh SK, Dua K, Kashyap MK. Epithelial-mesenchymal transition to mitigate age-related progression in lung cancer. Ageing Res Rev 2024; 102:102576. [PMID: 39515620 DOI: 10.1016/j.arr.2024.102576] [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: 09/05/2024] [Revised: 10/27/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Epithelial-Mesenchymal Transition (EMT) is a fundamental biological process involved in embryonic development, wound healing, and cancer progression. In lung cancer, EMT is a key regulator of invasion and metastasis, significantly contributing to the fatal progression of the disease. Age-related factors such as cellular senescence, chronic inflammation, and epigenetic alterations exacerbate EMT, accelerating lung cancer development in the elderly. This review describes the complex mechanism among EMT and age-related pathways, highlighting key regulators such as TGF-β, WNT/β-catenin, NOTCH, and Hedgehog signalling. We also discuss the mechanisms by which oxidative stress, mediated through pathways involving NRF2 and ROS, telomere attrition, regulated by telomerase activity and shelterin complex, and immune system dysregulation, driven by alterations in cytokine profiles and immune cell senescence, upregulate or downregulate EMT induction. Additionally, we highlighted pathways of transcription such as SNAIL, TWIST, ZEB, SIRT1, TP53, NF-κB, and miRNAs regulating these processes. Understanding these mechanisms, we highlight potential therapeutic interventions targeting these critical molecules and pathways.
Collapse
Affiliation(s)
- Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Saurabh Gupta
- Chameli Devi Institute of Pharmacy, Department of Pharmacology, Indore, Madhya Pradesh, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome-Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Smriti
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Madhav Singla
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Manoj Kumar Kashyap
- Molecular Oncology Laboratory, Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Panchgaon (Manesar), Gurugram, Haryana, India.
| |
Collapse
|
27
|
Tang Y, Shi T, Lin S, Fang T. Current status of research on the mechanisms of tumor-associated macrophages in esophageal cancer progression. Front Oncol 2024; 14:1450603. [PMID: 39678502 PMCID: PMC11638059 DOI: 10.3389/fonc.2024.1450603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/27/2024] [Indexed: 12/17/2024] Open
Abstract
Esophageal carcinoma (EC) is one of the most common tumors in China and seriously affects patient survival and quality of life. In recent years, increasing studies have shown that the tumor microenvironment is crucial in promoting tumor progression and metastasis. Tumor-associated macrophages (TAM) are key components of the tumor immune microenvironment and promote both tumor growth and antitumor immunity. Much evidence suggests that TAMs are closely associated with esophageal tumors. However, understanding of the clinical value and mechanism of action of TAM in esophageal cancer remains limited. Therefore, we reviewed the status of research on the role and mechanism of action of TAM in EC progression and summarized its potential clinical application value to provide a theoretical basis for the clinical treatment of EC.
Collapse
Affiliation(s)
- Yuchao Tang
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Tingting Shi
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- Group of Neuroendocrinology, Garvan Institute of Medical Research, Sydney, Australia
| | - Taiyong Fang
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
28
|
Cui Y, Zhang W, Zeng X, Yang Y, Park SJ, Nakai K. Computational analysis of the functional impact of MHC-II-expressing triple-negative breast cancer. Front Immunol 2024; 15:1497251. [PMID: 39664386 PMCID: PMC11631845 DOI: 10.3389/fimmu.2024.1497251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 11/08/2024] [Indexed: 12/13/2024] Open
Abstract
The tumor microenvironment (TME) plays a crucial role in tumor progression and immunoregulation. Major histocompatibility complex class II (MHC-II) is essential for immune surveillance within the TME. While MHC-II genes are typically expressed by professional antigen-presenting cells, they are also expressed in tumor cells, potentially facilitating antitumor immune responses. To understand the role of MHC-II-expressing tumor cells, we analyzed triple-negative breast cancer (TNBC), an aggressive subtype with poor prognosis and limited treatment options, using public bulk RNA-seq, single-cell RNA-seq, and spatial transcriptomics datasets. Our analysis revealed a distinct tumor subpopulation that upregulates MHC-II genes and actively interacts with immune cells. We implicated that this subpopulation is preferentially present in proximity to regions in immune infiltration of TNBC patient cohorts with a better prognosis, suggesting the functional importance of MHC-II-expressing tumor cells in modulating the immune landscape and influencing patient survival outcomes. Remarkably, we identified a prognostic signature comprising 40 significant genes in the MHC-II-expressing tumors in which machine leaning models with the signature successfully predicted patient survival outcomes and the degree of immune infiltration. This study advances our understanding of the immunological basis of cancer progression and suggests promising new directions for therapeutic strategies.
Collapse
Affiliation(s)
- Yang Cui
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Weihang Zhang
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Xin Zeng
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Yitao Yang
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Sung-Joon Park
- Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Kenta Nakai
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
- Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| |
Collapse
|
29
|
Miao L, Zhu Y, Chang H, Zhang X. Nanotheranostics in Breast Cancer Bone Metastasis: Advanced Research Progress and Future Perspectives. Pharmaceutics 2024; 16:1491. [PMID: 39771471 PMCID: PMC11676679 DOI: 10.3390/pharmaceutics16121491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/27/2024] [Accepted: 11/14/2024] [Indexed: 01/11/2025] Open
Abstract
Breast cancer is the leading cause of cancer-related morbidity and mortality among women worldwide, with bone being the most common site of all metastatic breast cancer. Bone metastases are often associated with pain and skeletal-related events (SREs), indicating poor prognosis and poor quality of life. Most current therapies for breast cancer bone metastasis primarily serve palliative purposes, focusing on pain management, mitigating the risk of bone-related complications, and inhibiting tumor progression. The emergence of nanodelivery systems offers novel insights and potential solutions for the diagnosis and treatment of breast cancer-related bone metastasis. This article reviews the recent advancements and innovative applications of nanodrug delivery systems in the context of breast cancer bone metastasis and explores future directions in nanotheranostics.
Collapse
Affiliation(s)
- Lin Miao
- Department of Breast Surgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang 110042, China; (L.M.); (Y.Z.)
- Graduate School, China Medical University, Shenyang 110122, China
| | - Yidan Zhu
- Department of Breast Surgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang 110042, China; (L.M.); (Y.Z.)
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
| | - Hong Chang
- Department of Breast Surgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang 110042, China; (L.M.); (Y.Z.)
| | - Xinfeng Zhang
- Department of Breast Surgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang 110042, China; (L.M.); (Y.Z.)
- Graduate School, China Medical University, Shenyang 110122, China
| |
Collapse
|
30
|
Xu J, Tang Z. Progress on angiogenic and antiangiogenic agents in the tumor microenvironment. Front Oncol 2024; 14:1491099. [PMID: 39629004 PMCID: PMC11611712 DOI: 10.3389/fonc.2024.1491099] [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: 09/04/2024] [Accepted: 10/31/2024] [Indexed: 12/06/2024] Open
Abstract
The development of tumors and their metastasis relies heavily on the process of angiogenesis. When the volume of a tumor expands, the resulting internal hypoxic conditions trigger the body to enhance the production of various angiogenic factors. These include vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), platelet-derived growth factor (PDGF), and transforming growth factor-α (TGF-α), all of which work together to stimulate the activation of endothelial cells and catalyze angiogenesis. Antiangiogenic therapy (AAT) aims to normalize tumor blood vessels by inhibiting these angiogenic signals. In this review, we will explore the molecular mechanisms of angiogenesis within the tumor microenvironment, discuss traditional antiangiogenic drugs along with their limitations, examine new antiangiogenic drugs and the advantages of combination therapy, and consider future research directions in the field of antiangiogenic drugs. This comprehensive overview aims to provide insights that may aid in the development of more effective anti-tumor treatments.
Collapse
Affiliation(s)
| | - Zhihua Tang
- Department of Pharmacy, Shaoxing People’s Hospital, Shaoxing, China
| |
Collapse
|
31
|
Lopes CDH, Braganca Xavier C, Torrado C, Veneziani AC, Megid TBC. A Comprehensive Exploration of Agents Targeting Tumor Microenvironment: Challenges and Future Perspectives. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2024; 7:283-299. [PMID: 39524466 PMCID: PMC11541921 DOI: 10.36401/jipo-24-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 08/13/2024] [Accepted: 08/20/2024] [Indexed: 11/16/2024]
Abstract
The tumor microenvironment (TME) encompasses the complex and diverse surroundings in which tumors arise. Emerging insights highlight the TME's critical role in tumor development, progression, metastasis, and treatment response. Consequently, the TME has attracted significant research and clinical interest, leading to the identification of numerous novel therapeutic targets. Advances in molecular technologies now enable detailed genomic and transcriptional analysis of cancer cells and the TME and the integration of microenvironmental data to the tumor genomic landscape. This comprehensive review discusses current progress in targeting the TME for drug development, addressing associated challenges, strategies for modulating the pro-tumor microenvironment, and the discovery of new targets.
Collapse
Affiliation(s)
| | | | - Carlos Torrado
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | |
Collapse
|
32
|
Wu X, Deng Y, Xu Y, Kang H, Hu JJ, Yoon J, Liang G. Activatable Fluorescence and Bio/Chemiluminescence Probes for Aminopeptidases: From Design to Biomedical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409893. [PMID: 39235570 DOI: 10.1002/adma.202409893] [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: 07/09/2024] [Revised: 08/27/2024] [Indexed: 09/06/2024]
Abstract
Aminopeptidases are exopeptidases that catalyze the cleavage of amino acid residues from the N-terminal fragment of protein or peptide substrates. Owing to their function, they play important roles in protein maturation, signal transduction, cell-cycle control, and various disease mechanisms, notably in cancer pathology. To gain better insights into their function, molecular imaging assisted by fluorescence and bio/chemiluminescence probes has become an indispensable method to their superiorities, including excellent sensitivity, selectivity, and real-time and noninvasive imaging. Numerous efforts are made to develop activatable probes that can effectively enhance efficiency and accuracy as well as minimize the side effects. This review is classified according to the type of aminopeptidases, summarizing some recent works on the design, work mechanism, and sensing, imaging, and theranostic performance of their activatable probe. Finally, the current challenges are outlined in developing activatable probes for aminopeptidases and provide possible solutions for future advancements.
Collapse
Affiliation(s)
- Xiaofeng Wu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yu Deng
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Ying Xu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, South Korea
| | - Jing-Jing Hu
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Juyoung Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, South Korea
| | - Gaolin Liang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| |
Collapse
|
33
|
Zhang M, He M, Bai L, Du F, Xie Y, Li B, Zhang Y. CircMALAT1 promotes the proliferation and metastasis of intrahepatic cholangiocarcinoma via the miR-512-5p/VCAM1 axis. Acta Biochim Biophys Sin (Shanghai) 2024; 57:223-236. [PMID: 39463204 PMCID: PMC11877140 DOI: 10.3724/abbs.2024185] [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: 12/25/2023] [Accepted: 05/06/2024] [Indexed: 10/29/2024] Open
Abstract
Circular RNAs play a pivotal role in the progression of various cancers. In our previous study, we observed high expression of the circRNA MALAT1 (cMALAT1) in intrahepatic cholangiocarcinoma (ICC) cells co-incubated with activated hepatic stellate cells. This study is designed to explore the roles of cMALAT1 and the underlying mechanisms in ICC. We find that cMALAT1 significantly facilitates the progression of ICC both in vitro and in vivo. The binding between cMALAT1 and miR-512-5p is subsequently confirmed through RNA pull-down experiments. As anticipated, the application of miR-512-5p mimics noticeably reverses the cMALAT1 overexpression-induced malignant phenotypes of ICC cells. Furthermore, VCAM1 is identified as a downstream gene of the cMALAT1/miR-512-5p axis. Importantly, silencing of VCAM1 not only effectively suppresses the malignant phenotypes of ICC cells but also significantly impairs the functions of cMALAT1. Our study reveals that cMALAT1 promotes the progression of ICC by competitively binding to VCAM1 mRNA with miR-512-5p, leading to the upregulation of VCAM1 expression and the activation of the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Meixia Zhang
- Department of Gastroenterologythe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
| | - Mingyan He
- Department of Gastroenterologythe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
| | - Liangliang Bai
- Department of Gastroenterologythe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
| | - Fan Du
- Department of Gastroenterologythe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
| | - Yingping Xie
- Department of Gastroenterologythe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
| | - Bimin Li
- Department of Gastroenterologythe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
| | - Yuming Zhang
- Department of SurgeryPeople’s Hospital of Nanchang Economic and Technological Development ZoneNanchang330013China
| |
Collapse
|
34
|
Santos da Silva T, da Silva-Júnior LN, Horvath-Pereira BDO, Valbão MCM, Garcia MHH, Lopes JB, Reis CHB, Barreto RDSN, Buchaim DV, Buchaim RL, Miglino MA. The Role of the Pancreatic Extracellular Matrix as a Tissue Engineering Support for the Bioartificial Pancreas. Biomimetics (Basel) 2024; 9:598. [PMID: 39451804 PMCID: PMC11505355 DOI: 10.3390/biomimetics9100598] [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: 07/31/2024] [Revised: 09/22/2024] [Accepted: 09/30/2024] [Indexed: 10/26/2024] Open
Abstract
Type 1 diabetes mellitus (T1DM) is a chronic condition primarily managed with insulin replacement, leading to significant treatment costs. Complications include vasculopathy, cardiovascular diseases, nephropathy, neuropathy, and reticulopathy. Pancreatic islet transplantation is an option but its success does not depend solely on adequate vascularization. The main limitations to clinical islet transplantation are the scarcity of human pancreas, the need for immunosuppression, and the inadequacy of the islet isolation process. Despite extensive research, T1DM remains a major global health issue. In 2015, diabetes affected approximately 415 million people, with projected expenditures of USD 1.7 trillion by 2030. Pancreas transplantation faces challenges due to limited organ availability and complex vascularization. T1DM is caused by the autoimmune destruction of insulin-producing pancreatic cells. Advances in biomaterials, particularly the extracellular matrix (ECM), show promise in tissue reconstruction and transplantation, offering structural and regulatory functions critical for cell migration, differentiation, and adhesion. Tissue engineering aims to create bioartificial pancreases integrating insulin-producing cells and suitable frameworks. This involves decellularization and recellularization techniques to develop biological scaffolds. The challenges include replicating the pancreas's intricate architecture and maintaining cell viability and functionality. Emerging technologies, such as 3D printing and advanced biomaterials, have shown potential in constructing bioartificial organs. ECM components, including collagens and glycoproteins, play essential roles in cell adhesion, migration, and differentiation. Clinical applications focus on developing functional scaffolds for transplantation, with ongoing research addressing immunological responses and long-term efficacy. Pancreatic bioengineering represents a promising avenue for T1DM treatment, requiring further research to ensure successful implementation.
Collapse
Affiliation(s)
- Thamires Santos da Silva
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (FMVZ/USP), São Paulo 05508-270, Brazil; (T.S.d.S.); (L.N.d.S.-J.); (B.d.O.H.-P.); (R.d.S.N.B.); (D.V.B.); (R.L.B.)
| | - Leandro Norberto da Silva-Júnior
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (FMVZ/USP), São Paulo 05508-270, Brazil; (T.S.d.S.); (L.N.d.S.-J.); (B.d.O.H.-P.); (R.d.S.N.B.); (D.V.B.); (R.L.B.)
- Postgraduate Department, University of Marília (UNIMAR), Marília 17525-902, Brazil; (M.C.M.V.); (M.H.H.G.); (J.B.L.)
| | - Bianca de Oliveira Horvath-Pereira
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (FMVZ/USP), São Paulo 05508-270, Brazil; (T.S.d.S.); (L.N.d.S.-J.); (B.d.O.H.-P.); (R.d.S.N.B.); (D.V.B.); (R.L.B.)
| | - Maria Carolina Miglino Valbão
- Postgraduate Department, University of Marília (UNIMAR), Marília 17525-902, Brazil; (M.C.M.V.); (M.H.H.G.); (J.B.L.)
| | | | - Juliana Barbosa Lopes
- Postgraduate Department, University of Marília (UNIMAR), Marília 17525-902, Brazil; (M.C.M.V.); (M.H.H.G.); (J.B.L.)
| | - Carlos Henrique Bertoni Reis
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia (UNIMAR), Marília 17525-902, Brazil;
- UNIMAR Beneficent Hospital (HBU), Medical School, University of Marilia (UNIMAR), Marilia 17525-160, Brazil
| | - Rodrigo da Silva Nunes Barreto
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (FMVZ/USP), São Paulo 05508-270, Brazil; (T.S.d.S.); (L.N.d.S.-J.); (B.d.O.H.-P.); (R.d.S.N.B.); (D.V.B.); (R.L.B.)
- Department of Animal Morphology and Physiology, Faculty of Agricultural and Veterinary Sciences, São Paulo State University, Jaboticabal 14884-900, Brazil
| | - Daniela Vieira Buchaim
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (FMVZ/USP), São Paulo 05508-270, Brazil; (T.S.d.S.); (L.N.d.S.-J.); (B.d.O.H.-P.); (R.d.S.N.B.); (D.V.B.); (R.L.B.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia (UNIMAR), Marília 17525-902, Brazil;
- Medical School, University Center of Adamantina (UNIFAI), Adamantina 17800-000, Brazil
| | - Rogerio Leone Buchaim
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (FMVZ/USP), São Paulo 05508-270, Brazil; (T.S.d.S.); (L.N.d.S.-J.); (B.d.O.H.-P.); (R.d.S.N.B.); (D.V.B.); (R.L.B.)
- Department of Biological Sciences, Bauru School of Dentistry (FOB/USP), University of Sao Paulo, Bauru 17012-901, Brazil
| | - Maria Angelica Miglino
- Postgraduate Department, University of Marília (UNIMAR), Marília 17525-902, Brazil; (M.C.M.V.); (M.H.H.G.); (J.B.L.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia (UNIMAR), Marília 17525-902, Brazil;
- Postgraduate Program in Animal Health, Production and Environment, University of Marilia (UNIMAR), Marilia 17525-902, Brazil
| |
Collapse
|
35
|
Liu Q, Yao F, Wu L, Xu T, Na J, Shen Z, Liu X, Shi W, Zhao Y, Liao Y. Heterogeneity and interplay: the multifaceted role of cancer-associated fibroblasts in the tumor and therapeutic strategies. Clin Transl Oncol 2024; 26:2395-2417. [PMID: 38602644 DOI: 10.1007/s12094-024-03492-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 03/31/2024] [Indexed: 04/12/2024]
Abstract
The journey of cancer development is a multifaceted and staged process. The array of treatments available for cancer varies significantly, dictated by the disease's type and stage. Cancer-associated fibroblasts (CAFs), prevalent across various cancer types and stages, play a pivotal role in tumor genesis, progression, metastasis, and drug resistance. The strategy of concurrently targeting cancer cells and CAFs holds great promise in cancer therapy. In this review, we focus intently on CAFs, delving into their critical role in cancer's progression. We begin by exploring the origins, classification, and surface markers of CAFs. Following this, we emphasize the key cytokines and signaling pathways involved in the interplay between cancer cells and CAFs and their influence on the tumor immune microenvironment. Additionally, we examine current therapeutic approaches targeting CAFs. This article underscores the multifarious roles of CAFs within the tumor microenvironment and their potential applications in cancer treatment, highlighting their importance as key targets in overcoming drug resistance and enhancing the efficacy of tumor therapies.
Collapse
Affiliation(s)
- Qiaoqiao Liu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Fei Yao
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Liangliang Wu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Tianyuan Xu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Jintong Na
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Zhen Shen
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Xiyu Liu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Wei Shi
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China.
- Department of Oncology, The First Affiliated Tumor Hospital, Guangxi University of Chinese Medicine, Nanning, 530021, Guangxi, China.
| | - Yongxiang Zhao
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China.
| | - Yuan Liao
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China.
| |
Collapse
|
36
|
Wang Y, Jia J, Wang F, Fang Y, Yang Y, Zhou Q, Yuan W, Gu X, Hu J, Yang S. Pre-metastatic niche: formation, characteristics and therapeutic implication. Signal Transduct Target Ther 2024; 9:236. [PMID: 39317708 PMCID: PMC11422510 DOI: 10.1038/s41392-024-01937-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/29/2024] [Accepted: 07/23/2024] [Indexed: 09/26/2024] Open
Abstract
Distant metastasis is a primary cause of mortality and contributes to poor surgical outcomes in cancer patients. Before the development of organ-specific metastasis, the formation of a pre-metastatic niche is pivotal in promoting the spread of cancer cells. This review delves into the intricate landscape of the pre-metastatic niche, focusing on the roles of tumor-derived secreted factors, extracellular vesicles, and circulating tumor cells in shaping the metastatic niche. The discussion encompasses cellular elements such as macrophages, neutrophils, bone marrow-derived suppressive cells, and T/B cells, in addition to molecular factors like secreted substances from tumors and extracellular vesicles, within the framework of pre-metastatic niche formation. Insights into the temporal mechanisms of pre-metastatic niche formation such as epithelial-mesenchymal transition, immunosuppression, extracellular matrix remodeling, metabolic reprogramming, vascular permeability and angiogenesis are provided. Furthermore, the landscape of pre-metastatic niche in different metastatic organs like lymph nodes, lungs, liver, brain, and bones is elucidated. Therapeutic approaches targeting the cellular and molecular components of pre-metastatic niche, as well as interventions targeting signaling pathways such as the TGF-β, VEGF, and MET pathways, are highlighted. This review aims to enhance our understanding of pre-metastatic niche dynamics and provide insights for developing effective therapeutic strategies to combat tumor metastasis.
Collapse
Affiliation(s)
- Yuhang Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Jiachi Jia
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Fuqi Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Yingshuai Fang
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Yabing Yang
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Quanbo Zhou
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Weitang Yuan
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Xiaoming Gu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China.
| | - Junhong Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China.
| | - Shuaixi Yang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China.
| |
Collapse
|
37
|
Andryszkiewicz W, Misiąg P, Karwowska A, Resler Z, Wojno A, Kulbacka J, Szewczyk A, Rembiałkowska N. Cancer Metastases to the Liver: Mechanisms of Tumor Cell Colonization. Pharmaceuticals (Basel) 2024; 17:1251. [PMID: 39338413 PMCID: PMC11434846 DOI: 10.3390/ph17091251] [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: 08/23/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
The liver is one of the most common sites for metastasis, which involves the spread from primary tumors to surrounding organs and tissues in the human body. There are a few steps in cancer expansion: invasion, inflammatory processes allowing the hepatic niche to be created, adhesions to ECM, neovascularization, and secretion of enzymes. The spread of tumor cells depends on the microenvironment created by the contribution of many biomolecules, including proteolytic enzymes, cytokines, growth factors, and cell adhesion molecules that enable tumor cells to interact with the microenvironment. Moreover, the microenvironment plays a significant role in tumor growth and expansion. The secreted enzymes help cancer cells facilitate newly formed hepatic niches and promote migration and invasion. Our study discusses pharmacological methods used to prevent liver metastasis by targeting the tumor microenvironment and cancer cell colonization in the liver. We examine randomized studies focusing on median survival duration and median overall survival in patients administered placebo compared with those treated with bevacizumab, ramucirumab, regorafenib, and ziv-aflibercept in addition to current chemotherapy. We also include research on mice and their responses to these medications, which may suppress metastasis progression. Finally, we discuss the significance of non-pharmacological methods, including surgical procedures, radiotherapy, cryotherapy, radiofrequency ablation (RFA), and transarterial embolization (TAE). In conclusion, the given methods can successfully prevent metastases to the liver and prolong the median survival duration and median overall survival in patients suffering from cancer.
Collapse
Affiliation(s)
- Wiktoria Andryszkiewicz
- The Students' Research Group, No. 148., Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland
| | - Piotr Misiąg
- The Students' Research Group, No. 148., Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland
| | - Anna Karwowska
- The Students' Research Group, No. 148., Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland
| | - Zofia Resler
- The Students' Research Group, No. 148., Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland
| | - Aleksandra Wojno
- The Students' Research Group, No. 148., Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Anna Szewczyk
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Nina Rembiałkowska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| |
Collapse
|
38
|
Jia J, Wang Y, Li M, Wang F, Peng Y, Hu J, Li Z, Bian Z, Yang S. Neutrophils in the premetastatic niche: key functions and therapeutic directions. Mol Cancer 2024; 23:200. [PMID: 39277750 PMCID: PMC11401288 DOI: 10.1186/s12943-024-02107-7] [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: 06/28/2024] [Accepted: 09/02/2024] [Indexed: 09/17/2024] Open
Abstract
Metastasis has been one of the primary reasons for the high mortality rates associated with tumours in recent years, rendering the treatment of current malignancies challenging and representing a significant cause of recurrence in patients who have undergone surgical tumour resection. Halting tumour metastasis has become an essential goal for achieving favourable prognoses following cancer treatment. In recent years, increasing clarity in understanding the mechanisms underlying metastasis has been achieved. The concept of premetastatic niches has gained widespread acceptance, which posits that tumour cells establish a unique microenvironment at distant sites prior to their migration, facilitating their settlement and growth at those locations. Neutrophils serve as crucial constituents of the premetastatic niche, actively shaping its microenvironmental characteristics, which include immunosuppression, inflammation, angiogenesis and extracellular matrix remodelling. These characteristics are intimately associated with the successful engraftment and subsequent progression of tumour cells. As our understanding of the role and significance of neutrophils in the premetastatic niche deepens, leveraging the presence of neutrophils within the premetastatic niche has gradually attracted the interest of researchers as a potential therapeutic target. The focal point of this review revolves around elucidating the involvement of neutrophils in the formation and shaping of the premetastatic niche (PMN), alongside the introduction of emerging therapeutic approaches aimed at impeding cancer metastasis.
Collapse
Affiliation(s)
- Jiachi Jia
- Zhengzhou University, Zhengzhou, 450000, China
| | - Yuhang Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Mengjia Li
- Department of Haematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Fuqi Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Yingnan Peng
- Department of Haematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Junhong Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Zhen Li
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China.
| | - Zhilei Bian
- Department of Haematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China.
| | - Shuaixi Yang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China.
| |
Collapse
|
39
|
Muñoz-Ayala A, Chimal-Vega B, Serafín-Higuera N, Galindo-Hernández O, Ramírez-Rosales G, Córdova-Guerrero I, Gómez-Lucas L, García-González V. Tamoxifen metabolites treatment promotes ERα+ transition to triple negative phenotype in vitro, effects of LDL in chemoresistance. Biosci Rep 2024; 44:BSR20240444. [PMID: 39034849 PMCID: PMC11301570 DOI: 10.1042/bsr20240444] [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: 04/15/2024] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024] Open
Abstract
OBJECTIVE Estrogen receptor-positive (ER+) breast cancer represents about 80% of cases, tamoxifen is the election neoadjuvant chemotherapy. However, a large percentage of patients develop chemoresistance, compromising recovery. Clinical evidence suggests that high plasmatic levels of low-density lipoproteins (LDL) could promote cancer progression. The present study analyzed the effect of LDL on the primary plasmatic active Tamoxifen's metabolites resistance acquisition, 4-hydroxytamoxifen (4OH-Tam) and 4-hydroxy-N-desmethyl-tamoxifen (endoxifen), in breast cancer ERα + cells (MCF-7). METHODS Two resistant cellular variants, MCF-7Var-H and MCF-7Var-I, were generated by a novel strategy and their phenotype features were evaluated. Phenotypic assessment was performed by MTT assays, cytometry, immunofluorescence microscopy, zymography and protein expression analysis. RESULTS MCF-7Var-H, generated only with tamoxifen metabolites, showed a critical down-regulation in hormone receptors, augmented migration capacity, metalloprotease 9 extracellular medium excretion, and a mesenchymal morphology in contrast with native MCF-7, suggesting the transition towards Triple-negative breast cancer (TNBC) phenotype. In contrast, MCF-7Var-I which was generated in a high LDL media, showed only a slight upregulation in ER and other less noticeable metabolic adaptations. Results suggest a potential role of transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) in phenotypic differences observed among variants. CONCLUSION LDL high or low concentrations during Tamoxifen´s metabolites chemoresistance acquisition leads to different cellular mechanisms related to chemoresistance. A novel adaptative cellular response associated with Nrf2 activity could be implicated.
Collapse
Affiliation(s)
- Andrea Muñoz-Ayala
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, México
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21000, México
| | - Brenda Chimal-Vega
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, México
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21000, México
| | - Nicolás Serafín-Higuera
- Facultad de Odontología Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, México
| | - Octavio Galindo-Hernández
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, México
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21000, México
| | - Gladys Ramírez-Rosales
- Departamento de Inmunología, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, México
| | - Iván Córdova-Guerrero
- Facultad de Ciencias Químicas e Ingeniería, Universidad Autónoma de Baja California, Tijuana 22390, México
| | - Luis Fernando Gómez-Lucas
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, México
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21000, México
| | - Victor García-González
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, México
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21000, México
| |
Collapse
|
40
|
Tomecka P, Kunachowicz D, Górczyńska J, Gebuza M, Kuźnicki J, Skinderowicz K, Choromańska A. Factors Determining Epithelial-Mesenchymal Transition in Cancer Progression. Int J Mol Sci 2024; 25:8972. [PMID: 39201656 PMCID: PMC11354349 DOI: 10.3390/ijms25168972] [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: 07/10/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 09/02/2024] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a process in which an epithelial cell undergoes multiple modifications, acquiring both morphological and functional characteristics of a mesenchymal cell. This dynamic process is initiated by various inducing signals that activate numerous signaling pathways, leading to the stimulation of transcription factors. EMT plays a significant role in cancer progression, such as metastasis and tumor heterogeneity, as well as in drug resistance. In this article, we studied molecular mechanisms, epigenetic regulation, and cellular plasticity of EMT, as well as microenvironmental factors influencing this process. We included both in vivo and in vitro models in EMT investigation and clinical implications of EMT, such as the use of EMT in curing oncological patients and targeting its use in therapies. Additionally, this review concludes with future directions and challenges in the wide field of EMT.
Collapse
Affiliation(s)
- Paulina Tomecka
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (P.T.); (J.G.); (M.G.); (J.K.); (K.S.)
| | - Dominika Kunachowicz
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211a, 50-556 Wroclaw, Poland;
| | - Julia Górczyńska
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (P.T.); (J.G.); (M.G.); (J.K.); (K.S.)
| | - Michał Gebuza
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (P.T.); (J.G.); (M.G.); (J.K.); (K.S.)
| | - Jacek Kuźnicki
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (P.T.); (J.G.); (M.G.); (J.K.); (K.S.)
| | - Katarzyna Skinderowicz
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (P.T.); (J.G.); (M.G.); (J.K.); (K.S.)
| | - Anna Choromańska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211a, 50-556 Wroclaw, Poland
| |
Collapse
|
41
|
Dong Y, Luo J, Pei M, Liu S, Gao Y, Zhou H, Nueraihemaiti Y, Zhan X, Xie T, Yao X, Guan X, Xu Y. Biomimetic Hydrogel-Mediated Mechano-Immunometabolic Therapy for Inhibition of ccRCC Recurrence After Surgery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308734. [PMID: 38884220 PMCID: PMC11321661 DOI: 10.1002/advs.202308734] [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: 11/14/2023] [Revised: 05/18/2024] [Indexed: 06/18/2024]
Abstract
The unique physical tumor microenvironment (TME) and aberrant immune metabolic status are two obstacles that must be overcome in cancer immunotherapy to improve clinical outcomes. Here, an in situ mechano-immunometabolic therapy involving the injection of a biomimetic hydrogel is presented with sequential release of the anti-fibrotic agent pirfenidone, which softens the stiff extracellular matrix, and small interfering RNA IDO1, which disrupts kynurenine-mediated immunosuppressive metabolic pathways, together with the multi-kinase inhibitor sorafenib, which induces immunogenic cell death. This combination synergistically augmented tumor immunogenicity and induced anti-tumor immunity. In mouse models of clear cell renal cell carcinoma, a single-dose peritumoral injection of a biomimetic hydrogel facilitated the perioperative TME toward a more immunostimulatory landscape, which prevented tumor relapse post-surgery and prolonged mouse survival. Additionally, the systemic anti-tumor surveillance effect induced by local treatment decreased lung metastasis by inhibiting epithelial-mesenchymal transition conversion. The versatile localized mechano-immunometabolic therapy can serve as a universal strategy for conferring efficient tumoricidal immunity in "cold" tumor postoperative interventions.
Collapse
Affiliation(s)
- Yunze Dong
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Jun Luo
- Department of UrologyShanghai Fourth People's HospitalSchool of MedicineTongji UniversityShanghai200434P. R. China
| | - Mingliang Pei
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025P. R. China
| | - Shuai Liu
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Yuchen Gao
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Hongmin Zhou
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Yimingniyizi Nueraihemaiti
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Xiangcheng Zhan
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Tiancheng Xie
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Xudong Yao
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Xin Guan
- Department of UltrasoundInstitute of Ultrasound in Medicine and EngineeringZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Yunfei Xu
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| |
Collapse
|
42
|
Chai Z, Zhu C, Wang X, Zheng Y, Han F, Xie Q, Liu C. PADI3 inhibits epithelial-mesenchymal transition by targeting CKS1-induced signal transduction in colon cancer. J Cancer Res Ther 2024; 20:1323-1333. [PMID: 39206995 DOI: 10.4103/jcrt.jcrt_558_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/03/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Protein arginine deiminase 3 (PADI3) is involved in various biological processes of human disease. PADI3 has recently received increasing attention due to its role in tumorigenesis. In a previous study, we found that PADI3 plays a tumor suppressor role in colon cancer by inducing cell cycle arrest, but its critical role and mechanism in cancer metastasis remain obscure. In this study, we fully studied the role of PADI3 in colon cancer cell metastasis. METHODS The expression levels of related proteins were detected by Western blotting, and Transwell and wound healing assays were used to examine the cell migration ability. Flow cytometry was used to measure and exclude cell apoptosis-affected cell migration. Both overexpression and rescue experiments were employed to elucidate the molecular mechanism of CKS1 in colon cancer cells. RESULTS The expression levels of PADI3 and CKS1 are negatively related, and PADI3 can promote CKS1 degradation in a ubiquitin-dependent manner. PADI3 can suppress colon cancer cell migration and reduce the wound healing speed by inhibiting CKS1 expression. The molecular mechanism showed that CKS1 can promote EMT by increasing Snail and N-cadherin expression and suppressing E-cadherin expression. PADI3, as a suppressor of CKS1, can block the process of EMT by impairing CKS1-induced Snail upregulation and E-cadherin downregulation; however, the expression of N-cadherin cannot be rescued. CONCLUSIONS CKS1 promotes EMT in colon cancer by regulating Snail/E-cadherin expression, and this effect can be reversed by PADI3 via the promotion of CKS1 degradation in a ubiquitylation-dependent manner.
Collapse
Affiliation(s)
- Zhengbin Chai
- Translational Research Laboratory for Stem Cell and Traditional Chinese Medicine, Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
- Department of Clinical Laboratory Medicine, Shandong Public Health Clinical Center, Shandong University, Jinan, People's Republic of China
| | - Changhui Zhu
- Department of Biochemistry and Molecular Biology, Shandong Provincial Qianfoshan Hospital, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, People's Republic of China
| | - Xiwei Wang
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| | - Yingying Zheng
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| | - Fabin Han
- Translational Research Laboratory for Stem Cell and Traditional Chinese Medicine, Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| | - Qi Xie
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| | - Chunyan Liu
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
- Jinan Clinical Research Center for Tissue Engineering Skin Regeneration and Wound Repair, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, People's Republic of China
- Department of Plastic Surgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, People's Republic of China
| |
Collapse
|
43
|
Guo Y, Ma S, Wang D, Mei F, Guo Y, Heng BC, Zhang S, Huang Y, Wei Y, He Y, Liu W, Xu M, Zhang X, Chen L, Deng X. HtrA3 paves the way for MSC migration and promotes osteogenesis. Bioact Mater 2024; 38:399-410. [PMID: 38774457 PMCID: PMC11107107 DOI: 10.1016/j.bioactmat.2024.05.016] [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: 01/24/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/24/2024] Open
Abstract
Mesenchymal stem cell (MSC) migration determines the healing capacity of bone and is crucial in promoting bone regeneration. Migration of MSCs is highly dependent on degradation of extracellular matrix by proteolytic enzymes. However, the underlying mechanisms of how enzymolysis paves the way for MSCs to migrate from their niche to the defect area is still not fully understood. Here, this study shows that high-temperature requirement A3 (HtrA3) overcomes the physical barrier and provides anchor points through collagen IV degradation, paving the way for MSC migration. HtrA3 is upregulated in MSCs at the leading edge of bone defect during the early stage of healing. HtrA3 degrades the surrounding collagen IV, which increases the collagen network porosity and increases integrin β1 expression. Subsequently, integrin β1 enhances the mechanotransduction of MSCs, thus remodeling the cytoskeleton, increasing cellular stiffness and nuclear translocation of YAP, eventually promoting the migration and subsequent osteogenic differentiation of MSCs. Local administration of recombinant HtrA3 in rat cranial bone defects significantly increases new bone formation and further validates the enhancement of MSC migration. This study helps to reveal the novel roles of HtrA3, explore potential targets for regenerative medicine, and offer new insights for the development of bioactive materials.
Collapse
Affiliation(s)
- Yaru Guo
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Siqin Ma
- Department of Stomatology, PLA General Hospital, First Affiliated Hospital (304 Hospital), Beijing, 100081, China
| | - Dandan Wang
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Feng Mei
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yusi Guo
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Boon Chin Heng
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Shihan Zhang
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Ying Huang
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Yan Wei
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Ying He
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Wenwen Liu
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Mingming Xu
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Xuehui Zhang
- NMPA Key Laboratory for Dental Materials, Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xuliang Deng
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- NMPA Key Laboratory for Dental Materials, Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| |
Collapse
|
44
|
Li Y, Wang M, Jiang L, Jia J, Pan F, Li W, Wang B, Huang K, Luo J. SIPA1 promotes epithelial-mesenchymal transition in colorectal cancer through STAT3 activation. Heliyon 2024; 10:e34527. [PMID: 39130435 PMCID: PMC11315193 DOI: 10.1016/j.heliyon.2024.e34527] [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: 01/05/2024] [Revised: 07/10/2024] [Accepted: 07/10/2024] [Indexed: 08/13/2024] Open
Abstract
Colorectal cancer (CRC) is the third leading cancer type worldwide and accounts for the second highest rate of cancer-related mortality. Liver metastasis significantly contributes to the mortality associated with CRC, but the fundamental mechanisms behind it remain unclear. Signal-induced proliferation-associated protein 1 (SIPA1), a GTPase activating protein, has been shown to promote metastasis in breast cancer. In this study, our objective was to explore the role of SIPA1 in regulating epithelial-mesenchymal transition (EMT) in CRC. The analysis of The Cancer Genome Atlas (TCGA) database revealed that the expression level of SIPA1 mRNA was notably upregulated and exhibited a positively correlated with EMT and STAT3 signaling pathways in CRC. Knockdown of SIPA1 impairs CRC cell proliferation and migration. Further studies on the reliance of SIPA1 on STAT3 signaling for EMT regulation have shown that SIPA1 stimulates the activation of STAT3, resulting in its nuclear translocation. The co-treatment of overexpressed SIPA1 with the STAT3 inhibitor STTITA has shown that SIPA1 regulates the expression of EMT-related markers through STAT3. Our study indicate that SIPA1 promotes CRC metastasis by activating the STAT3 signaling pathway, underscoring the potential of SIPA1 as a therapeutic target for metastatic CRC patients.
Collapse
Affiliation(s)
- Youjian Li
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, IATTI, Chongqing University of Arts and Sciences, Chongqing, China
| | - Mengjie Wang
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Jiang
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, IATTI, Chongqing University of Arts and Sciences, Chongqing, China
| | - Jiehong Jia
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, IATTI, Chongqing University of Arts and Sciences, Chongqing, China
| | - Fei Pan
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, IATTI, Chongqing University of Arts and Sciences, Chongqing, China
| | - Wen Li
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, IATTI, Chongqing University of Arts and Sciences, Chongqing, China
| | - Bochu Wang
- Biomedical and Health Engineering Laboratory, Chongqing University, Chongqing, China
| | - Ke Huang
- College of Landscape Architecture and Life Science/Institute of Special Plants, Chongqing University of Arts and Sciences, Yongchuan, Chongqing, China
| | - Jie Luo
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, IATTI, Chongqing University of Arts and Sciences, Chongqing, China
- Biomedical and Health Engineering Laboratory, Chongqing University, Chongqing, China
| |
Collapse
|
45
|
Zhang Y, Zheng Y, Zhang J, Xu C, Wu J. Apoptotic signaling pathways in bone metastatic lung cancer: a comprehensive analysis. Discov Oncol 2024; 15:310. [PMID: 39060849 PMCID: PMC11282049 DOI: 10.1007/s12672-024-01151-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
This review provides a comprehensive analysis of apoptotic signaling pathways in the context of bone metastatic lung cancer, emphasizing the intricate molecular mechanisms and microenvironmental influences. Beginning with an overview of apoptosis in cancer, the paper explores the specific molecular characteristics of bone metastatic lung cancer, highlighting alterations in apoptotic pathways. Focused discussions delve into key apoptotic signaling pathways, including the intrinsic and extrinsic pathways, and the roles of critical molecular players such as Bcl-2 family proteins and caspases. Microenvironmental factors, such as the tumor microenvironment, extracellular matrix interactions, and immune cell involvement, are examined in depth. The review also addresses experimental approaches and techniques employed in studying apoptotic signaling, paving the way for a discussion on current therapeutic strategies, their limitations, and future prospects. This synthesis contributes a holistic understanding of apoptosis in bone metastatic lung cancer, offering insights for potential therapeutic advancements.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Orthopedic Surgery, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
- Health Science Center, Ningbo University, Ningbo, 315211, Zhejiang, China
| | - Yi Zheng
- Department of Orthopedic Surgery, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Jiakai Zhang
- Department of Orthopedic Surgery, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Chaoyang Xu
- Hangzhou Medical College, Hangzhou, 310053, Zhejiang, China
| | - Junlong Wu
- Department of Orthopedic Surgery, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China.
| |
Collapse
|
46
|
You W, Azuma K, Iwagawa T, Watanabe S, Aihara M, Shiraya T, Ueta T. The role of lipid peroxidation in epithelial-mesenchymal transition of retinal pigment epithelial cells. Sci Rep 2024; 14:16498. [PMID: 39020017 PMCID: PMC11255318 DOI: 10.1038/s41598-024-67587-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024] Open
Abstract
Epithelial-Mesenchymal Transition (EMT) of retinal pigment epithelial (RPE) cells is recognized as pivotal in various retinal diseases. Previous studies have suggested a reciprocal regulation between reactive oxygen species (ROS) and EMT, though the involvement of peroxidized lipids or the effects of reducing them has remained unclear. The present study disclosed that EMT of ARPE-19 cells induced by TGF-β2 and TNF-α involves increased lipid peroxidation, and Ferrostatin-1 (Fer-1), a lipophilic antioxidative agent, successfully inhibited the increase in lipid peroxidation. Fer-1 suppressed the formation of EMT-associated fibrotic deposits, while EMT induction or Fer-1 treatment did not influence the cell viability or proliferation. Functionally, Fer-1 impeded EMT-driven cell migration and reduction in transepithelial electrical resistance. It demonstrated regulatory prowess by downregulating the mesenchymal marker fibronectin, upregulating the epithelial marker ZO-1, and inhibiting the EMT-associated transcriptional factor ZEB1. Additionally, VEGF, a major pathogenic cytokine in various retinal diseases, is also upregulated during EMT, and Fer-1 significantly mitigated the effect. The present study disclosed the involvement of lipid peroxidation in EMT of RPE cells, and suggests the suppression of lipid peroxidation may be a potential therapeutic target in retinal diseases in which EMT is implicated.
Collapse
Affiliation(s)
- Wang You
- Department of Ophthalmology, The Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| | - Kunihiro Azuma
- Department of Ophthalmology, The Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| | - Toshiro Iwagawa
- Department of Ophthalmology, The Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
- Department of Retinal Biology and Pathology, The Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| | - Sumiko Watanabe
- Department of Ophthalmology, The Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
- Department of Retinal Biology and Pathology, The Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| | - Makoto Aihara
- Department of Ophthalmology, The Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| | - Tomoyasu Shiraya
- Department of Ophthalmology, The Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan
| | - Takashi Ueta
- Department of Ophthalmology, The Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, Japan.
| |
Collapse
|
47
|
Wan H, Zhong L, Xia T, Zhang D. Silencing Exosomal circ102927 Inhibits Foot Melanoma Metastasis via Regulating Invasiveness, Epithelial-Mesenchymal Transition and Apoptosis. Cancer Manag Res 2024; 16:825-839. [PMID: 39044746 PMCID: PMC11263183 DOI: 10.2147/cmar.s460315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/09/2024] [Indexed: 07/25/2024] Open
Abstract
Background Exosomes contain abundant circular RNAs (circRNAs), playing an important role in intercellular communication. However, the function and underlying molecular mechanism of exosomal circRNAs in foot metastatic melanoma remain unclear. Methods Twelve differentially expressed exosomal circRNAs between patients with metastatic and primary foot melanoma were screened through high-throughput sequencing, and their expression levels were detected by the real-time reverse transcriptase-polymerase chain reaction (RT-qPCR). CircRNA102927 silencing and overexpression A2058 cell line was constructed, and the effects of circRNA102927 on cell proliferation, apoptosis, migration, invasion, and epithelial-mesenchymal transition (EMT) were assessed using cell counting kit-8 (CCK-8), flow cytometry, wound healing, Transwell, and Western blot assays, respectively. Results Twelve differentially expressed exosomal circRNAs were screened and ROC curve showed that six circRNAs could be used as the diagnostic biomarkers for metastatic melanoma. Melanoma-secreted exosomes induced the differentiation of CD4+ T cells into Treg cells. CircRNA102927 was highly expressed in metastatic melanomas. Functionally, circRNA102927 silencing inhibited proliferation, EMT, migration, and invasion in metastatic melanoma cells, while promoting apoptosis. Meanwhile, overexpression of circRNA102927 had the opposite effects. Conclusion Our investigation suggests that silencing exosomal circRNA102927 may suppress foot melanoma metastasis by inhibiting invasiveness, EMT and promoting apoptosis.
Collapse
Affiliation(s)
- Huiying Wan
- Department of Dermatology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Ling Zhong
- Department of Dermatology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Tian Xia
- Department of Pathology, Air Force Hospital of Western Theater Command, Chengdu, People’s Republic of China
| | - Dingding Zhang
- Sichuan Provincial Key Laboratory for Genetic Disease, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| |
Collapse
|
48
|
Long N, Sun RL, Lai QH, Lu MY, Li XH, Chen YN, Zhu DY. HOTAIR/miR-1277-5p/FBN2 signaling axis is involved in recurrent spontaneous abortion by regulating the growth, migration, and invasion of HTR-8/SVneo cells†. Biol Reprod 2024; 111:135-147. [PMID: 38401166 DOI: 10.1093/biolre/ioae030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/09/2024] [Accepted: 02/23/2024] [Indexed: 02/26/2024] Open
Abstract
OBJECTIVE This study aimed to explore the specific pathways by which HOX transcript antisense intergenic RNA contributes to the pathogenesis of unexplained recurrent spontaneous abortion. METHODS Real-time quantitative PCR was employed to assess the differential expression levels of HOX transcript antisense intergenic RNA in chorionic villi tissues from unexplained recurrent spontaneous abortion patients and women with voluntarily terminated pregnancies. HTR-8/SVneo served as a cellular model. Knockdown and overexpression of HOX transcript antisense intergenic RNA in the cells were achieved through siRNA transfection and pcDNA3.1 transfection, respectively. Cell viability, migration, and invasion were evaluated using cell counting kit-8, scratch, and Transwell assays, respectively. The interaction among the HOX transcript antisense intergenic RNA /miR-1277-5p/fibrillin 2 axis was predicted through bioinformatics analysis and confirmed through in vitro experiments. Furthermore, the regulatory effects of the HOX transcript antisense intergenic RNA /miR-1277-5p/fibrillin 2 signaling axis on cellular behaviors were validated in HTR-8/SVneo cells. RESULTS We found that HOX transcript antisense intergenic RNA was downregulated in chorionic villi tissues from unexplained recurrent spontaneous abortion patients. Overexpression of HOX transcript antisense intergenic RNA significantly enhanced the viability, migration, and invasion of HTR-8/SVneo cells, while knockdown of HOX transcript antisense intergenic RNA had the opposite effects. We further confirmed the regulatory effect of the HOX transcript antisense intergenic RNA /miR-1277-5p/fibrillin 2 signaling axis in unexplained recurrent spontaneous abortion. Specifically, HOX transcript antisense intergenic RNA and fibrillin 2 were found to reduce the risk of unexplained recurrent spontaneous abortion by enhancing cell viability, migration, and invasion, whereas miR-1277-5p exerted the opposite effects. CONCLUSION HOX transcript antisense intergenic RNA promotes unexplained recurrent spontaneous abortion development by targeting inhibition of miR-1277-5p/fibrillin 2 axis.
Collapse
Affiliation(s)
- Na Long
- Department of Reproductive Health, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, Guangdong, China
| | - Ru-Liang Sun
- Department of Pathology, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, Guangdong, China
| | - Qing-Hua Lai
- Department of Gynaecology, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, Guangdong, China
| | - Mei-Yin Lu
- Department of Sample Library, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, Guangdong, China
| | - Xiao-Hong Li
- Department of Reproductive Health, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, Guangdong, China
| | - Yan-Na Chen
- Department of Gynaecology, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, Guangdong, China
| | - Dong-Yan Zhu
- Department of Sample Library, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
49
|
Chen M, Shan H, Tao Q, Hu R, Sun Q, Zheng M, Chen Z, Lin Q, Yin M, Zhao S, Chen X, Chen Z. Mimicking Tumor Metastasis Using a Transwell-Integrated Organoids-On-a-Chip Platform. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308525. [PMID: 38308351 DOI: 10.1002/smll.202308525] [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: 09/25/2023] [Revised: 01/05/2024] [Indexed: 02/04/2024]
Abstract
The mortality rate among cancer patients is primarily attributed to tumor metastasis. The evaluation of metastasis potential provides a powerful framework for personalized therapies. However, little work has so far been undertaken to precisely model tumor metastasis in vitro, hindering the development of preventive and therapeutic interventions. In this work, a tumor-metastasis-mimicked Transwell-integrated organoids-on-a-chip platform (TOP) for precisely evaluating tumor metastatic potential is developed. Unlike the conventional Transwell device for detecting cell migration, the engineered device facilitates the assessment of metastasis in patient-derived organoids (PDO). Furthermore, a novel Transwell chamber with a hexagon-shaped structure is developed to mimic the migration of tumor cells into surrounding tissues, allowing for the evaluation of tumor metastasis in a horizontal direction. As a proof-of-concept demonstration, tumor organoids and metastatic clusters are further evaluated at the protein, genetic, and phenotypic levels. In addition, preliminary drug screening is undertaken to highlight the potential for using the device to combat cancers. In summary, the tumor-metastasis-mimicked TOP offers unique capabilities for evaluating the metastasis potential of tumor organoids and contributes to the development of personalized cancer therapies.
Collapse
Affiliation(s)
- Maike Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, China
- Furong Laboratory, Changsha, 410008, China
| | - Han Shan
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, China
- Furong Laboratory, Changsha, 410008, China
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, China
| | - Qian Tao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, China
| | - Rui Hu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, China
| | - Qi Sun
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, China
| | - Mingde Zheng
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, China
| | - Ziyan Chen
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, China
| | - Qibo Lin
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, China
| | - Mingzhu Yin
- Clinical Research Center (CRC), Medical Pathology Center (MPC), Cancer Early Detection and Treatment Center (CEDTC), Translational Medicine Research Center (TMRC), Chongqing University Three Gorges Hospital, Chongqing University, Chongqing, 404000, China
| | - Shuang Zhao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, China
- Furong Laboratory, Changsha, 410008, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Furong Laboratory, Changsha, 410008, China
| | - Zeyu Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, China
- Furong Laboratory, Changsha, 410008, China
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, China
| |
Collapse
|
50
|
Xu W, Ye J, Cao Z, Zhao Y, Zhu Y, Li L. Glucocorticoids in lung cancer: Navigating the balance between immunosuppression and therapeutic efficacy. Heliyon 2024; 10:e32357. [PMID: 39022002 PMCID: PMC11252876 DOI: 10.1016/j.heliyon.2024.e32357] [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: 03/22/2024] [Revised: 06/03/2024] [Accepted: 06/03/2024] [Indexed: 07/20/2024] Open
Abstract
Glucocorticoids (GCs), a class of hormones secreted by the adrenal glands, are released into the bloodstream to maintain homeostasis and modulate responses to various stressors. These hormones function by binding to the widely expressed GC receptor (GR), thereby regulating a wide range of pathophysiological processes, especially in metabolism and immunity. The role of GCs in the tumor immune microenvironment (TIME) of lung cancer (LC) has been a focal point of research. As immunosuppressive agents, GCs exert a crucial impact on the occurrence, progression, and treatment of LC. In the TIME of LC, GCs act as a constantly swinging pendulum, simultaneously offering tumor-suppressive properties while diminishing the efficacy of immune-based therapies. The present study reviews the role and mechanisms of GCs in the TIME of LC.
Collapse
Affiliation(s)
| | | | - Zhendong Cao
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, 210017, China
| | - Yupei Zhao
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, 210017, China
| | - Yimin Zhu
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, 210017, China
| | - Lei Li
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, 210017, China
| |
Collapse
|