1
|
Shu F, Wang Y, Li L, Shi L, Zhang F, Ma Z, Mao D. Multi-omics integration and machine learning identify and validate neutrophil extracellular trap-associated gene signatures in chronic rhinosinusitis with nasal polyps. Clin Immunol 2025; 275:110473. [PMID: 40089249 DOI: 10.1016/j.clim.2025.110473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
This study aimed to explore the molecular characteristics of neutrophil extracellular traps (NETs) in chronic rhinosinusitis with nasal polyps (CRSwNP). Differentially expressed gene analysis, weighted gene co-expression network analysis, and machine learning algorithms identified three core NETs-associated genes: CXCR4, CYBB, and PTAFR, which were significantly upregulated in CRSwNP patients. The diagnostic performance of these genes was evaluated using receiver operating characteristic (ROC) curves, and their clinical relevance was validated using multicenter data. Immune infiltration analysis showed strong correlations between these genes and neutrophil and immune cell infiltration. Single-cell RNA sequencing demonstrated that these genes were predominantly expressed in myeloid and immune cells and exhibited dynamic changes during disease progression. These genes may contribute to CRSwNP pathogenesis through IL-17 signaling and metabolism-related pathways. This study identifies novel biomarkers and therapeutic targets for precise diagnosis and personalized treatment of CRSwNP.
Collapse
Affiliation(s)
- Fu Shu
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400000, People's Republic of China
| | - Yaping Wang
- Department of Otorhinolaryngology, Yongchuan Chinese Medicine Hospital Affiliated to Chongqing Medical University, Chongqing 400000, People's Republic of China
| | - Linglong Li
- Department of Otorhinolaryngology, Yongchuan Chinese Medicine Hospital Affiliated to Chongqing Medical University, Chongqing 400000, People's Republic of China
| | - Lei Shi
- Department of Otorhinolaryngology, The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 110847, People's Republic of China
| | - Feng Zhang
- Department of Otorhinolaryngology, Yongchuan Chinese Medicine Hospital Affiliated to Chongqing Medical University, Chongqing 400000, People's Republic of China
| | - Zhixuan Ma
- Department of Otorhinolaryngology, The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 110847, People's Republic of China
| | - Dehong Mao
- Department of Otorhinolaryngology, Yongchuan Chinese Medicine Hospital Affiliated to Chongqing Medical University, Chongqing 400000, People's Republic of China.
| |
Collapse
|
2
|
Naz Z, Fareed M, Chaudhary ARH, Snigdha NT, Zafar A, Alsaidan OA, Mangu K, Ahmad S, Aslam M, Rizwanullah M. Exploring the therapeutic potential of ligand-decorated nanostructured lipid carriers for targeted solid tumor therapy. Int J Pharm 2025; 678:125687. [PMID: 40348302 DOI: 10.1016/j.ijpharm.2025.125687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/19/2025] [Accepted: 05/03/2025] [Indexed: 05/14/2025]
Abstract
Solid tumors present significant therapeutic challenges due to their complex pathophysiology, including poor vascularization, dense extracellular matrix, multidrug resistance, and immune evasion. Conventional treatment strategies, such as chemotherapy, radiotherapy, and surgical interventions, are often associated with systemic toxicity, suboptimal drug accumulation at the tumor site, and chemoresistance. Nanostructured lipid carriers (NLCs) have emerged as a promising approach to enhance anticancer therapy. NLCs offer several advantages, including high drug loading capacity, improved bioavailability, controlled release, and enhanced stability. Recent advancements in active targeting strategies have led to the development of ligand-decorated NLCs, which exhibit selective tumor targeting, improved cellular uptake, and reduced systemic toxicity. By functionalizing NLCs with different targeting ligands, site-specific drug delivery can be achieved for better therapeutic efficacy. This review comprehensively explores the potential of ligand-decorated NLCs in solid tumor therapy, highlights their design principles, and mechanisms of tumor targeting. Furthermore, it discusses various receptor-targeted NLCs for the effective treatment of solid tumors. The potential of ligand-decorated NLCs in combination therapy, gene therapy, photothermal therapy, and photodynamic therapy is also explored. Overall, ligand-decorated NLCs represent a versatile and effective strategy to achieve better therapeutic outcomes in solid tumor therapy.
Collapse
Affiliation(s)
- Zrien Naz
- Department of Pharmaceutics, College of Pharmacy, Al Asmarya University, Zliten 218521, Libya
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia
| | | | - Niher Tabassum Snigdha
- Department of Dental Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai 602105 Tamil Nadu, India
| | - Ameeduzzafar Zafar
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka-72341, Al-Jouf, Saudi Arabia
| | - Omar Awad Alsaidan
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka-72341, Al-Jouf, Saudi Arabia
| | - Karthik Mangu
- Kogniverse Education and Research, Bionest, Avishkaran (NIPER), Hyderabad-500037, Telangana, India
| | - Shahnawaz Ahmad
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohammed Aslam
- Pharmacy Department, Tishk International University, Erbil 44001 Kurdistan Region, Iraq
| | - Md Rizwanullah
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura 140401 Punjab, India.
| |
Collapse
|
3
|
Rueda A, Serna N, Mangues R, Villaverde A, Unzueta U. Targeting the chemokine receptor CXCR4 for cancer therapies. Biomark Res 2025; 13:68. [PMID: 40307933 PMCID: PMC12044942 DOI: 10.1186/s40364-025-00778-y] [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: 02/03/2025] [Accepted: 04/13/2025] [Indexed: 05/02/2025] Open
Abstract
The C-X-C chemokine receptor type 4 (CXCR4) has emerged as a key molecular biomarker for cancer therapies due to its critical role in tumor progression and metastases by displaying a stem cells phenotype. Its overexpression has been observed in more than 20 types of cancers, including solid tumors and hematological malignancies, and it is often associated with tumor aggressiveness and poor prognosis. Being initially recognized as a co-receptor involved in HIV infection, numerous CXCR4-targeting ligands and antagonists, including small molecules, peptides and biologics have been identified over the past decades. While only few of them have been used in the context of cancer therapies, recent biotechnological advancements using CXCR4 as a molecular target are showing significant potential to revolutionize future cancer therapies. Therefore, this review highlights the biotechnological innovations developed for cancer therapy and diagnosis by targeting the chemokine receptor CXCR4. It also discusses future perspectives on emerging therapeutic strategies, ranging from the use of small molecule inhibitors that block receptor signaling to cutting-edge nanocarriers designed for the targeted delivery of innovative drugs and proteins into cancer stem cells, aiming at cell-selective precision nanomedicines.
Collapse
Affiliation(s)
- Ariana Rueda
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77 - 79, Barcelona, 08041, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, 28029, Spain
- Josep Carreras Leukaemia Research Institute (IJC Sant Pau), 08041, Barcelona, Spain
| | - Naroa Serna
- CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, 28029, Spain
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Ramon Mangues
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77 - 79, Barcelona, 08041, Spain.
- CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, 28029, Spain.
- Josep Carreras Leukaemia Research Institute (IJC Sant Pau), 08041, Barcelona, Spain.
| | - Antonio Villaverde
- CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, 28029, Spain.
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193, Spain.
| | - Ugutz Unzueta
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77 - 79, Barcelona, 08041, Spain.
- CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, 28029, Spain.
- Josep Carreras Leukaemia Research Institute (IJC Sant Pau), 08041, Barcelona, Spain.
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193, Spain.
| |
Collapse
|
4
|
Cao Z, Wang Z, Yang L, Li T, Tao X, Niu X. Reshaping the immune microenvironment and reversing immunosenescence by natural products: Prospects for immunotherapy in gastric cancer. Semin Cancer Biol 2025; 110:1-16. [PMID: 39923925 DOI: 10.1016/j.semcancer.2025.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/25/2025] [Accepted: 02/03/2025] [Indexed: 02/11/2025]
Abstract
Gastric cancer (GC) represents a global health-care challenge. Recent progress in immunotherapy has elicited attracted considerable attention as a viable treatment option through modulating the host immune system and unleashing pre-existing immunity, which has profoundly revolutionized oncology, especially GC. Nonetheless, low clinical response and intrinsic and acquired resistance remain persistently challenging. The microenvironment of GC comprising multifarious stromal cell types has remarkable immunosuppressive elements that may impact the efficacy of immunotherapy. Immunosenescence is increasingly regarded as a factor that contributes to cancer development, remodels the tumor microenvironment and affects the efficacy of immunotherapy. Natural products are at the forefront of traditional medicine. Senotherapeutics is a class of drugs and natural products capable of delaying, preventing, or reversing the senescence process (i.e., senolytics) or suppressing senescence-associated secretory phenotype (i.e., senomorphics). Emerging evidence supports that natural products can improve the efficacy of existing immunotherapy and expand their indications in GC mainly based upon remodeling the immunosuppressive microenvironment and reversing immunosenescence. The review provides an integrated review of previously reported and ongoing clinical trials with immunotherapeutic regimens in GC and discusses current challenges. Next, we focus on natural compounds that exert anti-GC functions and possess immunomodulatory properties. More attention is paid to the potential of these natural compounds in modulating the immune microenvironment and immunosenescence. Lastly, we discuss the nanomedicine that can overcome the deficiencies of natural products. Altogether, our review suggests the enormous potential of natural compounds in GC immunotherapy, and provides an important direction for future research.
Collapse
Affiliation(s)
- Zhipeng Cao
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, China
| | - Zhilin Wang
- Department of Pain Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Li Yang
- Department of Anesthesiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Tian Li
- Tianjin Key Laboratory of Acute Abdomen Disease-Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China.
| | - Xueshu Tao
- Department of Pain Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | - Xing Niu
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
5
|
Huang H, Shi W, Yan H, Fan L, Lu J, Long Z, Li X, Li J, Wang J, Liu L, Qian J. Dual roles of CXCR4 (C-X-C motif chemokine receptor 4) in promoting entry of ebolavirus and targeting excessive glycoprotein for reticulophagic degradation to facilitate viral fitness. Autophagy 2025:1-20. [PMID: 40223186 DOI: 10.1080/15548627.2025.2492877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 04/15/2025] Open
Abstract
Ebola virus disease (EVD) caused by Zaire Ebolavirus (EBOV) infection is a major threat to public health in Africa and even worldwide, due to its extremely high mortality rate. However, there are still no effective antiviral therapies that can completely cure EVD. A comprehensive understanding of virus-host interactions would be beneficial for developing new antiviral agents. Here, we showed that CXCR4-induced macroautophagy/autophagy and was internalized to endosomes by interacting with glycoprotein (GP) on viral particles during EBOV infection; this promoted the EBOV attachment and entry, which was reduced by CXCR4 antagonist and neutralizing antibody. We also found that CXCR4 increased EBOV replication by downregulating cytotoxic GP to promote viral fitness instead of influencing the assembly of viral factory. Mechanistically, excessive EBOV GP could hijack CXCR4 sorting and transporting pathways by their interactions with HGS, one of the key components of the ESCRT machinery; subsequently GP could be carried back to the endoplasmic reticulum by CXCR4, where the E3 ubiquitin ligase RNF185 was recruited to polyubiquitinate GP in a K27- and K63-linked manner. Finally, polyubiquitinated GP was degraded in lysosomes via reticulophagy by interacting with RETREG1 (reticulophagy regulator 1), in an ATG3- and ATG5-dependent manner. Our findings revealed dual roles of CXCR4 in regulation of EBOV life cycle, either acting as an entry factor by interacting with GP on viral particles to facilitate viral entry or targeting excessive GP for reticulophagic degradation, providing new evidence that EBOV hijacked the host vesicular transportation system through efficient virus-host interactions to facilitate viral fitness.Abbreviations: Baf A1: bafilomycin A1; BDBV: Bundibugyo Ebolavirus; CHX: cycloheximide; CXCR4: C-X-C motif chemokine receptor 4; CLEC4M/DC-SIGNR: C type lectin domain family 4 member M; EBOV: Zaire Ebolavirus; EEA1: early endosome antigen 1; ER: endoplasmic reticulum; ERAD: ER-associated degradation; ESCRT: endosomal sorting complex required for transport; EVD: Ebolavirus disease; HAVCR1/TIM-1: hepatitis A virus cellular receptor 1; GP: glycoprotein; HGS: hepatocyte growth factor-regulated tyrosine kinase substrate; HIV: human immunodeficiency virus; IFL: internal fusion loop; ITCH/AIP4: itchy E3 ubiquitin protein ligase; LAMP: lysosomal associated membrane protein; LC-MS/MS: liquid chromatography mass spectrometry; PDIs: protein disulfide isomerases; RBD: receptor binding domain; RESTV: Reston Ebolavirus; RETREG1: reticulophagy regulator 1; RNF185: ring finger protein 185; SQSTM1/p62: sequestosome 1; SUDV: Sudan Ebolavirus; TAFV: Taï Forest Ebolavirus; TRIM21: tripartite motif containing 21; trVLPs: transcription- and replication-competent virus-like particles; Ub: ubiquitin.
Collapse
Affiliation(s)
- Hongxin Huang
- Department of Pathogen Biology and Biosecurity, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wendi Shi
- Department of Pathogen Biology and Biosecurity, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Huijun Yan
- Department of Pathogen Biology and Biosecurity, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Linjin Fan
- Department of Pathogen Biology and Biosecurity, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiajun Lu
- Department of Pathogen Biology and Biosecurity, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhenyu Long
- Institute of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaowei Li
- Department of Pathogen Biology and Biosecurity, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiao Li
- Department of Pathogen Biology and Biosecurity, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jie Wang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Linna Liu
- Institute of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jun Qian
- Department of Pathogen Biology and Biosecurity, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen, Guangdong, China
- Guangdong Provincial Highly Pathogenic Microorganism Science Data Center, Guangzhou, Guangdong, China
| |
Collapse
|
6
|
Shang T, Jia Z, Li J, Cao H, Xu H, Cong L, Ma D, Wang X, Liu J. Unraveling the triad of hypoxia, cancer cell stemness, and drug resistance. J Hematol Oncol 2025; 18:32. [PMID: 40102937 PMCID: PMC11921735 DOI: 10.1186/s13045-025-01684-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 03/05/2025] [Indexed: 03/20/2025] Open
Abstract
In the domain of addressing cancer resistance, challenges such as limited effectiveness and treatment resistance remain persistent. Hypoxia is a key feature of solid tumors and is strongly associated with poor prognosis in cancer patients. Another significant portion of the development of acquired drug resistance is attributed to tumor stemness. Cancer stem cells (CSCs), a small tumor cell subset with self-renewal and proliferative abilities, are crucial for tumor initiation, metastasis, and intra-tumoral heterogeneity. Studies have shown a significant association between hypoxia and CSCs in the context of tumor resistance. Recent studies reveal a strong link between hypoxia and tumor stemness, which together promote tumor survival and progression during treatment. This review elucidates the interplay between hypoxia and CSCs, as well as their correlation with resistance to therapeutic drugs. Targeting pivotal genes associated with hypoxia and stemness holds promise for the development of novel therapeutics to combat tumor resistance.
Collapse
Affiliation(s)
- Tongxuan Shang
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Ziqi Jia
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jiayi Li
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, 100730, China
| | - Heng Cao
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hengyi Xu
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lin Cong
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Dongxu Ma
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiang Wang
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jiaqi Liu
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
7
|
Zhao W, Sun J, Zhou B, Qiao H, Zhang J. Pan-cancer characterization of C-C motif chemokine ligand 5 (CCL5) identifies its role as biomarker and therapeutic target. Discov Oncol 2025; 16:264. [PMID: 40035930 PMCID: PMC11880496 DOI: 10.1007/s12672-025-02004-5] [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: 07/14/2024] [Accepted: 02/20/2025] [Indexed: 03/06/2025] Open
Abstract
OBJECTIVES C-C Motif Chemokine Ligand 5 (CCL5) is known for its role in immune regulation and has been implicated in cancer progression. However, its expression and prognostic significance in pan-cancer require comprehensive evaluation. This study was initiated to decipher the pan-cancer role of CCL5 genes. METHODS In silico analyses involving various online databases and molecular experiments involving CCL5 knockdown experiments in KIRC cell lines evaluated its role in cell proliferation, colony formation, and migration. RESULTS CCL5 expression was significantly up-regulated in several cancers. High CCL5 expression correlated with poorer overall survival in kidney renal cell carcinoma (KIRC) and esophageal cancer (ESCA) patients. Promoter hypomethylation correlated with elevated CCL5 expression and poorer prognosis. CCL5 mutations were rare; indicating its role in cancer is driven by overexpression rather than genetic alterations. Positive correlations with immune inhibitory and MHC genes suggested CCL5's role in fostering an immunosuppressive tumor microenvironment. High CCL5 expression correlated with increased immune cell infiltration, particularly CD8 T cells and macrophages. CCL5 expression did not significantly influence drug sensitivity. CCL5 knockdown in resulted in reduced proliferation, colony formation, and migration, underscoring its critical role in cancer cell dynamics. CONCLUSION Our study highlights the significance of CCL5 in cancer progression and prognosis, particularly in KIRC and ESCA. CCL5's role in modulating the tumor immune microenvironment and its potential as a therapeutic target warrant further investigation.
Collapse
Affiliation(s)
- Wenming Zhao
- Key Laboratory of Molecular Pathology and Early Diagnosis of Tumor in Hebei Province, Department of Pathology, Baoding First Central Hospital, Baoding, 071000, China
| | - Jirui Sun
- Key Laboratory of Molecular Pathology and Early Diagnosis of Tumor in Hebei Province, Department of Pathology, Baoding First Central Hospital, Baoding, 071000, China
| | - Bingjuan Zhou
- Key Laboratory of Molecular Pathology and Early Diagnosis of Tumor in Hebei Province, Department of Pathology, Baoding First Central Hospital, Baoding, 071000, China
| | - Haizhi Qiao
- Key Laboratory of Molecular Pathology and Early Diagnosis of Tumor in Hebei Province, Department of Pathology, Baoding First Central Hospital, Baoding, 071000, China
| | - Jinku Zhang
- Key Laboratory of Molecular Pathology and Early Diagnosis of Tumor in Hebei Province, Department of Pathology, Baoding First Central Hospital, Baoding, 071000, China.
| |
Collapse
|
8
|
Gao XQ, Li HL, Wang M, Yang CT, Su R, Shao LH. Kaempferol inhibited invasion and metastasis of gastric cancer cells by targeting AKT/GSK3β pathway based on network pharmacology and molecular docking. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2025; 27:421-441. [PMID: 39132822 DOI: 10.1080/10286020.2024.2387756] [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: 02/28/2024] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 08/13/2024]
Abstract
This study aims to explore the mechanisms of the inhibitory effect of kaempferol on the invasion and metastasis of gastric cancer (GC) cells through network pharmacology prediction and experimental verification. It identifies core targets via PPI network analysis and finds that kaempferol binds to these targets well. In vitro experiments showed that kaempferol could inhibit the proliferation, colony formation, migration and invasion of GC cells. Western blotting indicated kaempferol may reduce AKT and GSK3β phosphorylation, leading to lower expression of invasion-related genes SRC, MMP9, CXCR4, KDR, and MMP2. Overall, kaempferol may prevent migration and invasion of GC cells via the AKT/GSK3β signaling pathway.
Collapse
Affiliation(s)
- Xia-Qing Gao
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, China
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Hai-Long Li
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, China
- Department of Geriatrics, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Meng Wang
- Department of Geriatrics, Affiliated Hongqi Hospital of Mudanjiang Medical University, Mudanjiang 157011, China
- Research Center of Traditional Chinese Medicine, Gansu Province, Lanzhou 730000, China
| | - Chun-Ting Yang
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Rong Su
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Li-Hua Shao
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, China
| |
Collapse
|
9
|
Li T, Li X, Kang P, Zhao J. Exploring CX3CR1 as a prognostic biomarker and immunotherapeutic target in sarcoma. Transl Oncol 2025; 53:102283. [PMID: 39837057 PMCID: PMC11787715 DOI: 10.1016/j.tranon.2025.102283] [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: 11/08/2024] [Revised: 12/29/2024] [Accepted: 01/07/2025] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Sarcomas (SARC) are a diverse group of malignant tumors originating from mesenchymal tissues, characterized by poor prognosis under conventional therapies. CX3CR1, a chemokine receptor involved in immune cell migration, has emerged as a key player in SARC. Post-translational modifications (PTMs) such as phosphorylation and ubiquitination critically modulate CX3CR1, influencing cancer progression, immune responses, and treatment resistance. METHODS This study investigates CX3CR1 expression, its biological functions, and prognostic value in SARC. Using data from The Cancer Genome Atlas (TCGA), we analyzed CX3CR1 gene expression, methylation patterns, CRISPR screening results, and immune infiltration metrics. Functional experiments included knockout and overexpression models, CCK-8 assays and flow cytometry to assess apoptosis. RESULTS CX3CR1 expression was significantly elevated in SARC tissues and positively correlated with overall survival, disease-specific survival, and progression-free intervals. Methylation analysis identified CpG sites associated with CX3CR1 expression, differentiating tumor and adjacent tissues. CRISPR screening highlighted CX3CR1's essential role in tumor growth, while immune infiltration analysis underscored its impact on the tumor microenvironment. PTMs were found to stabilize CX3CR1, enhancing its activity in key signaling pathways. Overexpression of CX3CR1 amplified inflammatory and apoptotic responses, while knockdown showed protective effects in vitro. CONCLUSIONS CX3CR1 serves as a promising prognostic biomarker and therapeutic target in sarcoma. Targeting CX3CR1's PTMs could advance personalized treatments and improve outcomes for sarcoma patients.
Collapse
Affiliation(s)
- Tengfei Li
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xun Li
- Department of Orthopedics, Loudi Central Hospital, Ward 32, Loudi, China
| | - Pengcheng Kang
- Department of Orthopedics, Loudi Central Hospital, Ward 32, Loudi, China
| | - Jinmin Zhao
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| |
Collapse
|
10
|
Tang J, Wei W, Xu Y, Chen K, Miao Y, Fan W, Huang Z, Liu J, Chen P, Luo H, Wang L. CXC chemokine receptor 4 - mediated immune modulation and tumor microenvironment heterogeneity in gastric cancer: Utilizing multi-omics approaches to identify potential therapeutic targets. Biofactors 2025; 51:e2130. [PMID: 39431668 DOI: 10.1002/biof.2130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 09/29/2024] [Indexed: 10/22/2024]
Abstract
G-protein-coupled receptors (GPRs) are critical regulators of various biological behaviors, and their role in gastric cancer (GC) progression is gaining increasing attention. Among them, the immune regulatory mechanisms mediated by chemokine receptor 4 (CXCR4) remain insufficiently understood. This study aims to explore the immune regulatory functions of CXCR4 and the heterogeneity of the tumor microenvironment (TME) by examining GPR-related gene expression in GC. Through multi-omics approaches, including spatial transcriptomics and single-cell RNA sequencing, we investigated the oncogenic mechanisms of CXCR4, particularly its role in T cell immune exhaustion. In vitro experiments, including ELISA, PCR, CCK8 assays, cell scratch assays, and colony formation assays, were used to validate the role of CXCR4 in the migration and invasion of AGS and SNU-1 cell lines. CXCR4 silencing using siRNA further demonstrated its regulatory effects on these cellular processes. Our results revealed a strong correlation between elevated CXCR4 expression and increased exhaustion of regulatory T cells (Tregs) in the TME. Furthermore, heightened CXCR4 expression was linked to increased TME heterogeneity, driven by oxidative stress and activation of the NF-κB pathway, promoting immune evasion and tumor progression. Silencing CXCR4 significantly inhibited the invasive and proliferative abilities of AGS and SNU-1 cells, while also reducing the expression of pro-inflammatory cytokines IL-1β and interleukin-6, thus alleviating chronic inflammation and improving TME conditions. In conclusion, our comprehensive investigation highlights CXCR4 as a key mediator of TME dynamics and immune modulation in GC. Targeting CXCR4 presents a promising therapeutic strategy to slow tumor progression by reducing Tregs-mediated immune exhaustion and TME heterogeneity, positioning it as a novel therapeutic target in GC treatment.
Collapse
Affiliation(s)
- Jing Tang
- Department of Gastroenterology, Guangyuan Central Hospital, Guangyuan, China
| | - Wei Wei
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Yaoqing Xu
- Department of Clinical Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Department of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Kexin Chen
- Department of Clinical Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Department of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yaping Miao
- Department of Clinical Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Department of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Weining Fan
- Department of Clinical Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Department of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Zhi Huang
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China
| | - Jie Liu
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China
| | - Ping Chen
- Department of Clinical Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Department of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Honghao Luo
- Department of Radiology, Xichong People's Hospital, Nanchong, China
| | - Lexin Wang
- Department of Clinical Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Department of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
11
|
Chen N, Sun Y, Luo P, Tang Y, Fan Y, Han L, Wang K. Association of CXCR4 gene expression and promoter methylation with chronic hepatitis B-related fibrosis/cirrhosis. Int Immunopharmacol 2024; 139:112686. [PMID: 39053226 DOI: 10.1016/j.intimp.2024.112686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 06/30/2024] [Accepted: 07/11/2024] [Indexed: 07/27/2024]
Abstract
OBJECTIVE Chronic hepatitis B (CHB) virus infection remains a major public health concern. In this study, the diagnostic capability of C-X-C chemokine receptor type 4 promoter methylation in patients with CHB-associated liver fibrosis/cirrhosis was evaluated. METHODS Two hundred participants were recruited, including 25 healthy controls (HCs), 60 patients with CHB and 115 patients with hepatitis B virus (HBV)-related liver fibrosis/LC. Researchers monitored the methylation and messenger ribonucleic acid (mRNA) levels of C-X-C chemokine receptor type 4 (CXCR4) in peripheral blood mononuclear cells (PBMCs). In addition, we utilized single cell sequencing to analyze the cell types highly expressing CXCR4 in HBV-related liver fibrosis/LC. RESULTS HBV-related fibrosis/cirrhosis patients exhibited a significant elevation in the expression level of CXCR4 mRNA in PBMCs compared to CHB ones. The CXCR4 promoter showed a significantly lower methylation level in patients with CHB-related fibrosis/cirrhosis than in patients with CHB. Additionally, the diagnostic area under the area under the curve (AUC) of methylation of the CXCR4 promoter for CHB -related liver fibrosis/LC exceeded liver stiffness measurement (LSM), aspartate aminotransferase-to-platelet ratio index (APRI) and fibrosis-4 score (FIB-4). Furthermore, single-cell analysis demonstrated that CXCR4 expression is closely associated with Natural Killer cells(NK cells), T lymphocytes (T cells), and monocytes. CONCLUSION The low methylation of the CXCR4 promoter holds promise as a non-invasive biomarker for detecting CHB-associated liver fibrosis/LC.
Collapse
Affiliation(s)
- Nan Chen
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, PR China
| | - Yu Sun
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, PR China
| | - Pengyu Luo
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, PR China
| | - Yuna Tang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, PR China
| | - Yuchen Fan
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, PR China
| | - Liyan Han
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, PR China; Institute of Hepatology, Shandong University, Jinan 250012, PR China.
| | - Kai Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, PR China; Institute of Hepatology, Shandong University, Jinan 250012, PR China.
| |
Collapse
|
12
|
Yang J, Tian E, Chen L, Liu Z, Ren Y, Mao W, Zhang Y, Zhang J. Development and therapeutic perspectives of CXCR4 antagonists for disease therapy. Eur J Med Chem 2024; 275:116594. [PMID: 38879970 DOI: 10.1016/j.ejmech.2024.116594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 06/10/2024] [Accepted: 06/10/2024] [Indexed: 06/18/2024]
Abstract
Chemokine receptor 4 (CXCR4) is a subtype receptor protein of the GPCR family with a seven-transmembrane structure widely distributed in human tissues. CXCR4 is involved in diseases (e.g., HIV-1 infection), cancer proliferation and metastasis, inflammation signaling pathways, and leukemia, making it a promising drug target. Clinical trials on CXCR4 antagonists mainly focused on peptides and antibodies, with a few small molecule compounds, such as AMD11070 (2) and MSX-122 (3), showing promise in cancer treatment. This perspective discusses the structure-activity relationship (SAR) of CXCR4 and its role in diseases, mainly focusing on the SAR of CXCR4 antagonists. It also explores the standard structural features and target interactions of CXCR4 binding in different disease categories. Furthermore, it investigates various modification strategies to propose potential improvements in the effectiveness of CXCR4 drugs.
Collapse
Affiliation(s)
- Jun Yang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Erkang Tian
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Li Chen
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zihang Liu
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yijiu Ren
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Wuyu Mao
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Yiwen Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Jifa Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center and Institute of Respiratory Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
13
|
Wei X, Wu F, Dong H, Jing Y, Song Y, Pang H, Chen J, Du Z, He W, Ma L, Wang Y, Hu J, Li Q, Yang S. 68Ga-pentixafor PET/CT in the localization diagnosis of primary aldosteronism concurrent subclinical cushing's syndrsome: two case reports. Endocrine 2024; 85:1398-1406. [PMID: 38914747 DOI: 10.1007/s12020-024-03865-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 11/28/2023] [Indexed: 06/26/2024]
Abstract
PURPOSE Adrenal venous sampling (AVS) is recommended for subtyping primary aldosteronism (PA). However, in cases of PA, concurrent subclinical Cushing's syndrome (SCS) has the potential to confound AVS results. Pentixafor, a CXC chemokine receptor type 4-specific ligand, has been reported as a promising marker to evaluate functional nature of adrenal adenomas. This study aims to investigate the clinical value of Gallium-68 Pentixafor Positron Emission Tomography-Computed Tomography (68Ga-Pentixafor PET/CT) in the localization diagnosis of patients with PA plus SCS. METHODS Two patients with a confirmed diagnosis of PA plus SCS underwent AVS and 68Ga-Pentixafor PET/CT. RESULTS AVS results revealed no lateralization for both patients while 68Ga-Pentixafor PET/CT showed a unilateral adrenal nodule with increased uptake of 68Ga-Pentixafor. Unilateral adrenalectomy was performed based on the results of 68Ga-Pentixafor PET/CT. Subsequently, complete biochemical remission of autonomous aldosterone and cortisol secretion were achieved in both cases. CONCLUSIONS 68Ga-Pentixafor PET/CT shows promising potential for the localization of aldosterone and cortisol co-secreting adrenal adenoma in patients with PA plus SCS.
Collapse
Affiliation(s)
- Xin Wei
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Feifei Wu
- Department of Endocrinology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Haoyu Dong
- Department of Endocrinology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Ying Jing
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ying Song
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hua Pang
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Chen
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhipeng Du
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wenwen He
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Linqiang Ma
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Wang
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jinbo Hu
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qifu Li
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Shumin Yang
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
14
|
Sergio I, Varricchio C, Patel SK, Del Gaizo M, Russo E, Orlando A, Peruzzi G, Ferrandino F, Tsaouli G, Coni S, Peluso D, Besharat ZM, Campolo F, Venneri MA, Del Bufalo D, Lai S, Indraccolo S, Minuzzo S, La Starza R, Bernardini G, Screpanti I, Campese AF, Felli MP. Notch3-regulated microRNAs impair CXCR4-dependent maturation of thymocytes allowing maintenance and progression of T-ALL. Oncogene 2024; 43:2535-2547. [PMID: 38907003 DOI: 10.1038/s41388-024-03079-0] [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: 07/13/2023] [Revised: 05/30/2024] [Accepted: 06/05/2024] [Indexed: 06/23/2024]
Abstract
Malignant transformation of T-cell progenitors causes T-cell acute lymphoblastic leukemia (T-ALL), an aggressive childhood lymphoproliferative disorder. Activating mutations of Notch, Notch1 and Notch3, have been detected in T-ALL patients. In this study, we aimed to deeply characterize hyperactive Notch3-related pathways involved in T-cell dynamics within the thymus and bone marrow to propose these processes as an important step in facilitating the progression of T-ALL. We previously generated a transgenic T-ALL mouse model (N3-ICtg) demonstrating that aberrant Notch3 signaling affects early thymocyte maturation programs and leads to bone marrow infiltration by CD4+CD8+ (DP) T cells that are notably, Notch3highCXCR4high. Newly, our in vivo results suggest that an anomalous immature thymocyte subpopulation, such as CD4-CD8- (DN) over-expressing CD3ɛ, but with low CXCR4 expression, dominates N3-ICtg thymus-resident DN subset in T-ALL progression. MicroRNAs might be of significance in T-ALL pathobiology, however, whether required for leukemia maintenance is not fully understood. The selection of specific DN subsets demonstrates the inverse correlation between CXCR4 expression and a panel of Notch3-deregulated miRNAs. Interestingly, we found that within DN thymocyte subset hyperactive Notch3 inhibits CXCR4 expression through the cooperative effects of miR-139-5p and miR-150-5p, thus impinging on thymocyte differentiation with accumulation of DNCD3ɛ+CXCR4- cells. These data point out that deregulation of Notch3 in T-ALL, besides its role in sustaining dissemination of abnormal DP T cells, as we previously demonstrated, could play a role in selecting specific DN immature T cells within the thymus, thus impeding T cell development, to facilitate T-ALL progression inside the bone marrow.
Collapse
Affiliation(s)
- Ilaria Sergio
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Claudia Varricchio
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Martina Del Gaizo
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Eleonora Russo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Andrea Orlando
- Division of Hematology & Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York NY, USA
| | | | | | - Georgia Tsaouli
- Department of Medical-Surgical Science and Translational Medicine, Sapienza University of Rome, Rome, Italy
| | - Sonia Coni
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Daniele Peluso
- Ph.D School of Applied Medical-Surgical Sciences, Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | | | - Federica Campolo
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Donatella Del Bufalo
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Silvia Lai
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Stefano Indraccolo
- Department of Surgery Oncology and Gastroenterology, University of Padua, Padua, Italy
- Basic and Translational Oncology Unit, Istituto Oncologico Veneto IOV-IRCCS, Padua, Italy
| | - Sonia Minuzzo
- Department of Surgery Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Roberta La Starza
- Hematology Section, Department of Medicine and Surgery, and S. Maria Della Misericordia Hospital Perugia, CREO, Perugia, Italy
| | | | - Isabella Screpanti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Maria Pia Felli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
15
|
Yang Y, Yuan L, Liu W, Lu D, Meng F, Yang Y, Zhou Z, Ma P, Nan Y. Banxia-Shengjiang drug pair inhibits gastric cancer development and progression by improving body immunity. Medicine (Baltimore) 2024; 103:e36303. [PMID: 38457601 PMCID: PMC10919495 DOI: 10.1097/md.0000000000036303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 03/10/2024] Open
Abstract
To investigate the mechanism of action of Banxia-Shengjiang drug pair on the inhibition of gastric cancer (GC) using network pharmacology and bioinformatics techniques. The action targets of the Banxia (Pinellia ternata (Thunb.) Makino) -Shengjiang (Zingiber officinale Roscoe) drug pair obtained from the TCMSP database were intersected with differentially expressed genes (DEGs) and GC-related genes, and the intersected genes were analyzed for pathway enrichment to identify the signaling pathways and core target genes. Subsequently, the core target genes were analyzed for clinical relevance gene mutation analysis, methylation analysis, immune infiltration analysis and immune cell analysis. Finally, by constructing the PPI network of hub genes and corresponding active ingredients, the key active ingredients of the Banxia-Shengjiang drug pair were screened for molecular docking with the hub genes. In this study, a total of 557 target genes of Banxia-Shengjiang pairs, 7754 GC-related genes and 1799 DEGs in GC were screened. Five hub genes were screened, which were PTGS2, MMP9, PPARG, MMP2, and CXCR4. The pathway enrichment analyses showed that the intersecting genes were associated with RAS/MAPK signaling pathway. In addition, the clinical correlation analysis showed that hub genes were differentially expressed in GC and was closely associated with immune infiltration and immunotherapy. The results of single nucleotide variation (SNV) and copy number variation (CNV) indicated that mutations in the hub genes were associated with the survival of gastric cancer patients. Finally, the PPI network and molecular docking results showed that PTGS2 and MMP9 were potentially important targets for the inhibition of GC by Banxia-Shengjiang drug pair, while cavidine was an important active ingredient for the inhibition of GC by Banxia-Shengjiang drug pair. Banxia-Shengjiang drug pair may regulate the immune function and inhibit GC by modulating the expression of core target genes such as RAS/MAPK signaling pathway, PTGS2 and MMP9.
Collapse
Affiliation(s)
- Yating Yang
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Ling Yuan
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Wenjing Liu
- Key Laboratory of Hui Ethnic Medicine Modernization of Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Doudou Lu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Fandi Meng
- Key Laboratory of Hui Ethnic Medicine Modernization of Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yi Yang
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Ziying Zhou
- Pharmacy Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Ping Ma
- Pharmacy Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yi Nan
- Key Laboratory of Hui Ethnic Medicine Modernization of Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
16
|
Borges J, Zeng J, Liu XQ, Chang H, Monge C, Garot C, Ren K, Machillot P, Vrana NE, Lavalle P, Akagi T, Matsusaki M, Ji J, Akashi M, Mano JF, Gribova V, Picart C. Recent Developments in Layer-by-Layer Assembly for Drug Delivery and Tissue Engineering Applications. Adv Healthc Mater 2024; 13:e2302713. [PMID: 38116714 PMCID: PMC11469081 DOI: 10.1002/adhm.202302713] [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: 08/17/2023] [Revised: 11/27/2023] [Indexed: 12/21/2023]
Abstract
Surfaces with biological functionalities are of great interest for biomaterials, tissue engineering, biophysics, and for controlling biological processes. The layer-by-layer (LbL) assembly is a highly versatile methodology introduced 30 years ago, which consists of assembling complementary polyelectrolytes or biomolecules in a stepwise manner to form thin self-assembled films. In view of its simplicity, compatibility with biological molecules, and adaptability to any kind of supporting material carrier, this technology has undergone major developments over the past decades. Specific applications have emerged in different biomedical fields owing to the possibility to load or immobilize biomolecules with preserved bioactivity, to use an extremely broad range of biomolecules and supporting carriers, and to modify the film's mechanical properties via crosslinking. In this review, the focus is on the recent developments regarding LbL films formed as 2D or 3D objects for applications in drug delivery and tissue engineering. Possible applications in the fields of vaccinology, 3D biomimetic tissue models, as well as bone and cardiovascular tissue engineering are highlighted. In addition, the most recent technological developments in the field of film construction, such as high-content liquid handling or machine learning, which are expected to open new perspectives in the future developments of LbL, are presented.
Collapse
Grants
- GA259370 ERC "BIOMIM"
- GA692924 ERC "BioactiveCoatings"
- GA790435 ERC "Regenerbone"
- ANR-17-CE13-022 Agence Nationale de la Recherche "CODECIDE", "OBOE", "BuccaVac"
- ANR-18-CE17-0016 Agence Nationale de la Recherche "CODECIDE", "OBOE", "BuccaVac"
- 192974 Agence Nationale de la Recherche "CODECIDE", "OBOE", "BuccaVac"
- ANR-20-CE19-022 BIOFISS Agence Nationale de la Recherche "CODECIDE", "OBOE", "BuccaVac"
- ANR22-CE19-0024 SAFEST Agence Nationale de la Recherche "CODECIDE", "OBOE", "BuccaVac"
- DOS0062033/0 FUI-BPI France
- 883370 European Research Council "REBORN"
- 2020.00758.CEECIND Portuguese Foundation for Science and Technology
- UIDB/50011/2020,UIDP/50011/2020,LA/P/0006/2020 FCT/MCTES (PIDDAC)
- 751061 European Union's Horizon 2020 "PolyVac"
- 11623 Sidaction
- 20H00665 JSPS Grant-in-Aid for Scientific Research
- 3981662 BPI France Aide Deep Tech programme
- ECTZ60600 Agence Nationale de Recherches sur le Sida et les Hépatites Virales
- 101079482 HORIZON EUROPE Framework Programme "SUPRALIFE"
- 101058554 Horizon Europe EIC Accelerator "SPARTHACUS"
- Sidaction
- Agence Nationale de Recherches sur le Sida et les Hépatites Virales
Collapse
Affiliation(s)
- João Borges
- CICECO – Aveiro Institute of MaterialsDepartment of ChemistryUniversity of AveiroCampus Universitário de SantiagoAveiro3810‐193Portugal
| | - Jinfeng Zeng
- Division of Applied ChemistryGraduate School of EngineeringOsaka University2‐1 YamadaokaSuitaOsaka565–0871Japan
| | - Xi Qiu Liu
- School of PharmacyTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Hao Chang
- Hangzhou Institute of MedicineChinese Academy of SciencesHangzhouZhejiang310022China
| | - Claire Monge
- Laboratory of Tissue Biology and Therapeutic Engineering (LBTI)UMR5305 CNRS/Universite Claude Bernard Lyon 17 Passage du VercorsLyon69367France
| | - Charlotte Garot
- Université de Grenoble AlpesCEAINSERM U1292 BiosantéCNRS EMR 5000 Biomimetism and Regenerative Medicine (BRM)17 avenue des MartyrsGrenobleF‐38054France
| | - Ke‐feng Ren
- Department of Polymer Science and EngineeringZhejiang UniversityHangzhou310027China
| | - Paul Machillot
- Université de Grenoble AlpesCEAINSERM U1292 BiosantéCNRS EMR 5000 Biomimetism and Regenerative Medicine (BRM)17 avenue des MartyrsGrenobleF‐38054France
| | - Nihal E. Vrana
- SPARTHA Medical1 Rue Eugène BoeckelStrasbourg67000France
| | - Philippe Lavalle
- SPARTHA Medical1 Rue Eugène BoeckelStrasbourg67000France
- Institut National de la Santé et de la Recherche MédicaleInserm UMR_S 1121 Biomaterials and BioengineeringCentre de Recherche en Biomédecine de Strasbourg1 rue Eugène BoeckelStrasbourg67000France
- Université de StrasbourgFaculté de Chirurgie Dentaire1 place de l'HôpitalStrasbourg67000France
| | - Takami Akagi
- Building Block Science Joint Research ChairGraduate School of Frontier BiosciencesOsaka University1–3 YamadaokaSuitaOsaka565–0871Japan
| | - Michiya Matsusaki
- Division of Applied ChemistryGraduate School of EngineeringOsaka University2‐1 YamadaokaSuitaOsaka565–0871Japan
| | - Jian Ji
- Department of Polymer Science and EngineeringZhejiang UniversityHangzhou310027China
| | - Mitsuru Akashi
- Building Block Science Joint Research ChairGraduate School of Frontier BiosciencesOsaka University1–3 YamadaokaSuitaOsaka565–0871Japan
| | - João F. Mano
- CICECO – Aveiro Institute of MaterialsDepartment of ChemistryUniversity of AveiroCampus Universitário de SantiagoAveiro3810‐193Portugal
| | - Varvara Gribova
- Institut National de la Santé et de la Recherche MédicaleInserm UMR_S 1121 Biomaterials and BioengineeringCentre de Recherche en Biomédecine de Strasbourg1 rue Eugène BoeckelStrasbourg67000France
- Université de StrasbourgFaculté de Chirurgie Dentaire1 place de l'HôpitalStrasbourg67000France
| | - Catherine Picart
- Université de Grenoble AlpesCEAINSERM U1292 BiosantéCNRS EMR 5000 Biomimetism and Regenerative Medicine (BRM)17 avenue des MartyrsGrenobleF‐38054France
| |
Collapse
|
17
|
Xu H, Chen S, Li J, Weng S, Ren Y, Zhang Y, Wang L, Liu L, Guo C, Xing Z, Luo P, Cheng Q, Han X, Liu Z. Cellular Ligand-Receptor Perturbations Unravel MEIS2 as a Key Factor for the Aggressive Progression and Prognosis in Stage II/III Colorectal Cancer. J Proteome Res 2024; 23:760-774. [PMID: 38153233 DOI: 10.1021/acs.jproteome.3c00626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Approximately 10-15% of stage II and 25-30% of stage III colorectal cancer (CRC) patients experience recurrence within 5 years after surgery, and existing taxonomies are insufficient to meet the needs of clinical precision treatment. Thus, robust biomarkers and precise management were urgently required to stratify stage II and III CRC and identify potential patients who will benefit from postoperative adjuvant therapy. Alongside, interactions of ligand-receptor pairs point to an emerging direction in tumor signaling with far-reaching implications for CRC, while their impact on tumor subtyping has not been elucidated. Herein, based on multiple large-sample multicenter cohorts and perturbations of the ligand-receptor interaction network, four well-characterized ligand-receptor-driven subtypes (LRDS) were established and further validated. These molecular taxonomies perform with unique heterogeneity in terms of molecular characteristics, immune and mutational landscapes, and clinical features. Specifically, MEIS2, a key LRDS4 factor, performs significant associations with proliferation, invasion, migration, and dismal prognosis of stage II/III CRC, revealing promising directions for prognostic assessment and individualized treatment of CRC patients. Overall, our study sheds novel insights into the implications of intercellular communication on stage II/III CRC from a ligand-receptor interactome perspective and revealed MEIS2 as a key factor in the aggressive progression and prognosis for stage II/III CRC.
Collapse
Affiliation(s)
- Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China
| | - Shuang Chen
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jing Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China
| | - Libo Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Long Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Chunguang Guo
- Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zhe Xing
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P. R. China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
18
|
Anastasiadou DP, Quesnel A, Duran CL, Filippou PS, Karagiannis GS. An emerging paradigm of CXCL12 involvement in the metastatic cascade. Cytokine Growth Factor Rev 2024; 75:12-30. [PMID: 37949685 DOI: 10.1016/j.cytogfr.2023.10.003] [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: 10/15/2023] [Accepted: 10/20/2023] [Indexed: 11/12/2023]
Abstract
The chemokine CXCL12, also known as stromal cell-derived factor 1 (SDF1), has emerged as a pivotal regulator in the intricate molecular networks driving cancer progression. As an influential factor in the tumor microenvironment, CXCL12 plays a multifaceted role that spans beyond its traditional role as a chemokine inducing invasion and metastasis. Indeed, CXCL12 has been assigned functions related to epithelial-to-mesenchymal transition, cancer cell stemness, angiogenesis, and immunosuppression, all of which are currently viewed as specialized biological programs contributing to the "metastatic cascade" among other cancer hallmarks. Its interaction with its cognate receptor, CXCR4, initiates a cascade of events that not only shapes the metastatic potential of tumor cells but also defines the niches within the secondary organs that support metastatic colonization. Given the profound implications of CXCL12 in the metastatic cascade, understanding its mechanistic underpinnings is of paramount importance for the targeted elimination of rate-limiting steps in the metastatic process. This review aims to provide a comprehensive overview of the current knowledge surrounding the role of CXCL12 in cancer metastasis, especially its molecular interactions rationalizing its potential as a therapeutic target.
Collapse
Affiliation(s)
- Dimitra P Anastasiadou
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA; Tumor Microenvironment & Metastasis Program, Albert Einstein Cancer Center, Bronx, NY, USA
| | - Agathe Quesnel
- School of Health & Life Sciences, Teesside University, Middlesbrough TS1 3BX, United Kingdom; National Horizons Centre, Teesside University, Darlington DL1 1HG, United Kingdom
| | - Camille L Duran
- Tumor Microenvironment & Metastasis Program, Albert Einstein Cancer Center, Bronx, NY, USA; Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA; Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Panagiota S Filippou
- School of Health & Life Sciences, Teesside University, Middlesbrough TS1 3BX, United Kingdom; National Horizons Centre, Teesside University, Darlington DL1 1HG, United Kingdom
| | - George S Karagiannis
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA; Tumor Microenvironment & Metastasis Program, Albert Einstein Cancer Center, Bronx, NY, USA; Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA; Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
19
|
Liu W. The Involvement of Cysteine-X-Cysteine Motif Chemokine Receptors in Skin Homeostasis and the Pathogenesis of Allergic Contact Dermatitis and Psoriasis. Int J Mol Sci 2024; 25:1005. [PMID: 38256077 PMCID: PMC10815665 DOI: 10.3390/ijms25021005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Members of the C-X-C motif chemokine receptor (CXCR) superfamily play central roles in initiating the innate immune response in mammalian cells by orchestrating selective cell migration and immune cell activation. With its multilayered structure, the skin, which is the largest organ in the body, performs a crucial defense function, protecting the human body from harmful environmental threats and pathogens. CXCRs contribute to primary immunological defense; these receptors are differentially expressed by different types of skin cells and act as key players in initiating downstream innate immune responses. While the initiation of inflammatory responses by CXCRs is essential for pathogen elimination and tissue healing, overactivation of these receptors can enhance T-cell-mediated autoimmune responses, resulting in excessive inflammation and the development of several skin disorders, including psoriasis, atopic dermatitis, allergic contact dermatitis, vitiligo, autoimmune diseases, and skin cancers. In summary, CXCRs serve as critical links that connect innate immunity and adaptive immunity. In this article, we present the current knowledge about the functions of CXCRs in the homeostasis function of the skin and their contributions to the pathogenesis of allergic contact dermatitis and psoriasis. Furthermore, we will examine the research progress and efficacy of therapeutic approaches that target CXCRs.
Collapse
Affiliation(s)
- Wenjie Liu
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| |
Collapse
|
20
|
Jiang Y, Gao Y, Li X, He F, Liu Y, Wang R. Caulis Spatholobi extracts inhibit osteosarcoma growth and metastasis through suppression of CXCR4/PI3K/AKT signaling. J Orthop Surg Res 2023; 18:985. [PMID: 38129870 PMCID: PMC10740265 DOI: 10.1186/s13018-023-04437-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 12/01/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND The therapeutic potential of Caulis Spatholobi (CS) extracts against various cancers has been well documented, yet its impact and mechanism in osteosarcoma (OS) remain unexplored. This study aims to elucidate the effects of CS extracts on the growth and metastasis of OS, along with its underlying molecular mechanism. METHODS The impact of CS extracts on the proliferative potential of two OS cell lines (Saos-2 and U2OS) was assessed using MTT and colony-formation assays. Additionally, the migratory and invasive capacities of OS cells were investigated through Transwell assays. The modulation of CXCR4 expression by CS extracts was evaluated using qRT-PCR and Western blotting. Furthermore, the influence of CS extracts on the activation of PI3K/Akt signaling was determined through Western blotting. RESULTS CS extracts exhibited a dose- and time-dependent inhibition of proliferation and colony formation in OS cells. Notably, CXCR4 expression was prominently observed in Saos-2 and U2OS, and treatment with CS extracts led to a dose-dependently reduction in CXCR4 levels. Silencing CXCR4 or inhibiting its function diminished the migratory and invasive capacities of OS cells. Conversely, the CS extracts induced suppression of OS cell migration and invasion was counteracted by CXCR4 overexpression. Mechanistically, CS extracts repressed PI3K/AKT signaling in OS cells by downregulating CXCR4 expression. CONCLUSIONS CS extracts mitigate the CXCR4/PI3K/AKT signaling-mediated growth and metastasis capacities of OS cells, thus might play an anti-tumor role in OS.
Collapse
Affiliation(s)
- Yang Jiang
- Traditional Medicine Department, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Yemei Gao
- Traditional Medicine Department, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Xin Li
- Traditional Medicine Department, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Fangming He
- Traditional Medicine Department, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Yang Liu
- Traditional Medicine Department, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Renxian Wang
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, 31 Xinjiekou East Street, Xicheng District, Beijing, 100035, China.
| |
Collapse
|
21
|
Sellner F, Compérat E, Klimpfinger M. Genetic and Epigenetic Characteristics in Isolated Pancreatic Metastases of Clear-Cell Renal Cell Carcinoma. Int J Mol Sci 2023; 24:16292. [PMID: 38003482 PMCID: PMC10671160 DOI: 10.3390/ijms242216292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/09/2023] [Accepted: 11/11/2023] [Indexed: 11/26/2023] Open
Abstract
Isolated pancreatic metastases of renal cell carcinoma (IsPMRCC) are a rare manifestation of metastatic, clear-cell renal cell carcinoma (RCC) in which distant metastases occur exclusively in the pancreas. In addition to the main symptom of the isolated occurrence of pancreatic metastases, the entity surprises with additional clinical peculiarities: (a) the unusually long interval of about 9 years between the primary RCC and the onset of pancreatic metastases; (b) multiple pancreatic metastases occurring in 36% of cases; (c) favourable treatment outcomes with a 75% 5-year survival rate; and (d) volume and growth-rate dependent risk factors generally accepted to be relevant for overall survival in metastatic surgery are insignificant in isPMRCC. The genetic and epigenetic causes of exclusive pancreatic involvement have not yet been investigated and are currently unknown. Conversely, according to the few available data in the literature, the following genetic and epigenetic peculiarities can already be identified as the cause of the protracted course: 1. high genetic stability of the tumour cell clones in both the primary tumour and the pancreatic metastases; 2. a low frequency of copy number variants associated with aggressiveness, such as 9p, 14q and 4q loss; 3. in the chromatin-modifying genes, a decreased rate of PAB1 (3%) and an increased rate of PBRM1 (77%) defects are seen, a profile associated with a favourable course; 4. an increased incidence of KDM5C mutations, which, in common with increased PBRM1 alterations, is also associated with a favourable outcome; and 5. angiogenetic biomarkers are increased in tumour tissue, while inflammatory biomarkers are decreased, which explains the good response to TKI therapy and lack of sensitivity to IT.
Collapse
Affiliation(s)
- Franz Sellner
- Department of General, Visceral and Vascular Surgery, Clinic Favoriten Vienna, Kaiser Franz Josef Hospital, 1100 Vienna, Austria
| | - Eva Compérat
- Clinical Institute of Pathology, Medical University Vienna, 1090 Vienna, Austria
| | - Martin Klimpfinger
- Clinical Institute of Pathology, Medical University Vienna, 1090 Vienna, Austria
| |
Collapse
|
22
|
Zhang S, Shen T, Zeng Y. Epigenetic Modifications in Prostate Cancer Metastasis and Microenvironment. Cancers (Basel) 2023; 15:cancers15082243. [PMID: 37190171 DOI: 10.3390/cancers15082243] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/30/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023] Open
Abstract
The gradual evolution of prostate tissue from benign tumor to malignant lesion or distant metastasis is driven by intracellular epigenetic changes and the tumor microenvironment remodeling. With the continuous study of epigenetic modifications, these tumor-driving forces are being discovered and are providing new treatments for cancer. Here we introduce the classification of epigenetic modification and highlight the role of epigenetic modification in tumor remodeling and communication of the tumor microenvironment.
Collapse
Affiliation(s)
- Shouyi Zhang
- Department of Urology, the Cancer Hospital of Dalian University of Technology & Liaoning Cancer Hospital, Shenyang 110042, China
| | - Tao Shen
- Department of Urology, Second Affiliated Hospital of Shenyang Medical College, No. 20 Beijiu Road, Heping District, Shenyang 110001, China
| | - Yu Zeng
- Department of Urology, the Cancer Hospital of Dalian University of Technology & Liaoning Cancer Hospital, Shenyang 110042, China
| |
Collapse
|
23
|
Siddiqui JA, Nasser MW. Editorial: Role of chemokines in tumor heterogeneity. Semin Cancer Biol 2023; 92:128-129. [PMID: 37028577 DOI: 10.1016/j.semcancer.2023.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
|
24
|
Liu G, Chen T, Zhang X, Ma X, Shi H. Small molecule inhibitors targeting the cancers. MedComm (Beijing) 2022; 3:e181. [PMID: 36254250 PMCID: PMC9560750 DOI: 10.1002/mco2.181] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/23/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022] Open
Abstract
Compared with traditional therapies, targeted therapy has merits in selectivity, efficacy, and tolerability. Small molecule inhibitors are one of the primary targeted therapies for cancer. Due to their advantages in a wide range of targets, convenient medication, and the ability to penetrate into the central nervous system, many efforts have been devoted to developing more small molecule inhibitors. To date, 88 small molecule inhibitors have been approved by the United States Food and Drug Administration to treat cancers. Despite remarkable progress, small molecule inhibitors in cancer treatment still face many obstacles, such as low response rate, short duration of response, toxicity, biomarkers, and resistance. To better promote the development of small molecule inhibitors targeting cancers, we comprehensively reviewed small molecule inhibitors involved in all the approved agents and pivotal drug candidates in clinical trials arranged by the signaling pathways and the classification of small molecule inhibitors. We discussed lessons learned from the development of these agents, the proper strategies to overcome resistance arising from different mechanisms, and combination therapies concerned with small molecule inhibitors. Through our review, we hoped to provide insights and perspectives for the research and development of small molecule inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Gui‐Hong Liu
- Department of BiotherapyState Key Laboratory of BiotherapyCancer Center, West China HospitalSichuan UniversityChengduChina
| | - Tao Chen
- Department of CardiologyThe First Affiliated Hospital of China Medical UniversityShenyangLiaoningChina
| | - Xin Zhang
- Department of BiotherapyState Key Laboratory of BiotherapyCancer Center, West China HospitalSichuan UniversityChengduChina
| | - Xue‐Lei Ma
- Department of BiotherapyState Key Laboratory of BiotherapyCancer Center, West China HospitalSichuan UniversityChengduChina
| | - Hua‐Shan Shi
- Department of BiotherapyState Key Laboratory of BiotherapyCancer Center, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
25
|
Ahmad A. Exosomes in Cancer Diagnosis and Therapy. Int J Mol Sci 2022; 23:ijms23179930. [PMID: 36077326 PMCID: PMC9456038 DOI: 10.3390/ijms23179930] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Affiliation(s)
- Aamir Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| |
Collapse
|
26
|
Shen Y, Lu M, Xu Q, Liu L, Cheng Z. Effect of Bairui Granule on Inflammatory Mediators in Induced Sputum, Leukotriene C4, and EOS in Peripheral Blood of Children with Cough Variant Asthma. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:2657994. [PMID: 35720028 PMCID: PMC9200585 DOI: 10.1155/2022/2657994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 04/24/2022] [Accepted: 04/29/2022] [Indexed: 11/18/2022]
Abstract
Objective To study the effect of Hanchuan Zupa granule combined with conventional western medicine in the treatment of children with bronchial asthma. Methods 98 cases in Fengrun District People's Hospital of Tangshan City from June 2018 to February 2021 were selected. The control group was given oxygen therapy, antibiotics, and aerosol inhalation of quick acting β 2 receptor agonist, glucocorticoid, and other conventional western medicine treatment, while the observation group was treated with Bairui granule on the basis of the control group. The course of treatment of the two groups was 1 week. Results After treatment, the levels of sputum IL-4, IL-17, neu, and ECP in the two groups decreased, and the observation group was lower than the control group (P < 0.05). The levels of EOS, CXCR4, LTB4, and SDF-1 in peripheral blood of the two groups were lower than those in the control group (P < 0.05). The daytime cough, night cough, and TCM syndrome scores of the two groups were decreased, and the observation group was lower than the control group (P < 0.05). Conclusion Bairui granule combined with conventional western medicine in the treatment of children with bronchial asthma, the curative effect is worthy of affirmation, can effectively improve cough symptoms, reduce EOS, CXCR, LTB4, SDF-1 levels, inhibit airway inflammation, and has good clinical application value.
Collapse
Affiliation(s)
- Yi Shen
- Department of Paediatrics, The Affiliated Suzhou Science &Technology Town Hospital of Nanjing Medical University, China
| | - Min Lu
- Department of Paediatrics, The Affiliated Suzhou Science &Technology Town Hospital of Nanjing Medical University, China
| | - Qiuyan Xu
- Department of Paediatrics, The Affiliated Suzhou Science &Technology Town Hospital of Nanjing Medical University, China
| | - Lu Liu
- Department of Paediatrics, The Affiliated Suzhou Science &Technology Town Hospital of Nanjing Medical University, China
| | - Zhi Cheng
- Department of Paediatrics, The Affiliated Suzhou Science &Technology Town Hospital of Nanjing Medical University, China
| |
Collapse
|