1
|
Law YY, Tran NB, Song CY, Wu YY, Chen HT, Fong YC, Tsai HC, Kuo YH, Tang CH. Antcin K inhibits chondrosarcoma motility by reducing MMP‑7 expression via downregulation of the PI3K, Akt, mTOR and NF‑κB signaling pathway. Mol Med Rep 2025; 32:180. [PMID: 40280107 PMCID: PMC12056539 DOI: 10.3892/mmr.2025.13545] [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: 11/08/2024] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
Chondrosarcoma is the second most common form of primary bone cancer originating from cartilage. Chondrosarcoma cells have a high propensity to spread to other organs during the advanced stage, with the lung being a preferred site. Although surgery is the most effective treatment for chondrosarcoma, it has low efficacy in the metastasis stage. Antrodia cinnamomea is the source of the triterpenoid antcin K, which exhibits immunomodulatory and anti‑inflammatory properties. However, the therapeutic function of antcin K on chondrosarcoma has not yet been elucidated. The inhibitory effect of antcin K was evaluated using migration and invasion assays while cell toxicity was determined using the MTT assay. Molecular function regulation by antcin K was investigated by RNA sequencing and Ingenuity Pathway Analysis. The present study revealed that antcin K decreases migration and invasion in two chondrosarcoma cell lines. RNA sequencing revealed that MMP‑7 serves a key role in antcin K‑mediated motility of chondrosarcoma cells. Antcin K diminished MMP‑7 expression, and overexpression of MMP‑7 antagonized antcin K‑induced inhibition of cell migration and invasion. Antcin K abolished the activation of PI3K, Akt, mTOR and NF‑κB pathways. The present study demonstrated that antcin K is a novel candidate for chondrosarcoma motility inhibition by decreasing the PI3K, Akt, mTOR and NF‑κB signaling cascades, which inhibits MMP‑7 production.
Collapse
Affiliation(s)
- Yat-Yin Law
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan 40201, Taiwan, R.O.C
- Department of Orthopedics, Chung Shan Medical University Hospital, Taichung, Taiwan 402, Taiwan, R.O.C
| | - Nguyen Bao Tran
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan 404, Taiwan, R.O.C
| | - Chang-Yu Song
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan 404, Taiwan, R.O.C
| | - Yu-Ying Wu
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan 40201, Taiwan, R.O.C
- Department of Orthopedics, Chung Shan Medical University Hospital, Taichung, Taiwan 402, Taiwan, R.O.C
- Department of Orthopedics, Penghu Hospital, Ministry of Health and Welfare, Penghu, Taiwan 880, Taiwan, R.O.C
| | - Hsien-Te Chen
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan 404, Taiwan, R.O.C
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan 404, Taiwan, R.O.C
| | - Yi-Chin Fong
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan 404, Taiwan, R.O.C
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan 404, Taiwan, R.O.C
- Department of Orthopedic Surgery, China Medical University Beigang Hospital, Taichung, Yunlin, Taiwan 651, Taiwan, R.O.C
| | - Hsiao-Chi Tsai
- Department of Medicine Research, China Medical University Beigang Hospital, Yunlin, Taiwan 651, Taiwan, R.O.C
| | - Yueh-Hsiung Kuo
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan 404, Taiwan, R.O.C
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan 404333, Taiwan, R.O.C
| | - Chih-Hsin Tang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan 404, Taiwan, R.O.C
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan 404333, Taiwan, R.O.C
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan 404, Taiwan, R.O.C
- Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan 41354, Taiwan, R.O.C
| |
Collapse
|
2
|
Sinha S, Roy R, Barman N, Sarkar P, Saha A, Biswas N. IL6 mediated cFLIP downregulation increases the migratory and invasive potential of triple negative breast cancer cell. Cell Signal 2025; 130:111679. [PMID: 39988287 DOI: 10.1016/j.cellsig.2025.111679] [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: 08/14/2024] [Revised: 02/03/2025] [Accepted: 02/16/2025] [Indexed: 02/25/2025]
Abstract
c-FLIP (cellular FLICE-Like Inhibitor of Apoptotic protein) alias CFLAR (Cellular FADD-like apoptosis regulator) is an inhibitor of Caspase 8 and thus plays a key role in the regulation of extrinsic apoptotic pathway. However, the mechanisms of cFLIP regulation during the course of progression of cancer and it's involvement in tumour cell migration and invasion is yet to be known. Our TCGA data analysis has shown that cFLIP is downregulated in many cancers, including breast cancer, especially at the later stages. Next, we have analysed the role of cFLIP in breast cancer progression in In-vitro study. In doing so, we have used luminal breast cancer cell line MCF7 as non-aggressive and non-invasive breast cancer model and triple negative breast cancer cell lines MDA-MB-231, MDA-MB-468 and MDA-MB-453 as highly aggressive and invasive breast cancer cell model. When, we analysed and compared MCF7 and triple negative cell lines, we found a negative correlation between cFLIP expression pattern and metastasis which supported our In-silico study. Moreover, we found that Il6, one of the most prominent cytokines inside tumour microenvironment, helped in cFLIP downregulation via activation of p38 in MDA-MB-231 cell line. Not only that we have shown that cFLIP negatively regulated autophagy and this autophagy down-regulation resulted in decrease in metastasis. Thus, we have shown in an In-vitro model, for the first time, a complete interconnecting pathway in which IL6 mediated p38 activation directly influences metastasis by regulating autophagy via cFLIP downregulation.
Collapse
Affiliation(s)
- Samraj Sinha
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Rajdeep Roy
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Nilesh Barman
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Purandar Sarkar
- Institute of Health Sciences, Presidency University, Kolkata, India
| | - Abhik Saha
- Institute of Health Sciences, Presidency University, Kolkata, India
| | - Nabendu Biswas
- Department of Life Sciences, Presidency University, Kolkata, India.
| |
Collapse
|
3
|
Siddiqui AJ, Patel M, Jahan S, Abdelgadir A, Alam MJ, Alshahrani MM, Alturaiki W, Sachidanandan M, Khan A, Badraoui R, Adnan M. Silver Nanoparticles Derived from Probiotic Lactobacillus casei-a Novel Approach for Combating Bacterial Infections and Cancer. Probiotics Antimicrob Proteins 2025; 17:1277-1294. [PMID: 38085438 DOI: 10.1007/s12602-023-10201-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2023] [Indexed: 05/07/2025]
Abstract
In the face of rising antibiotic resistance and the need for novel therapeutic approaches against cancer, the present study delves into the various facets of biosynthesized silver nanoparticles (AgNPs) derived from the probiotic strain Lactobacillus casei (AgNPs-LC), assessing their efficacy in combating bacterial infections, disrupting biofilm formation, interfering with quorum sensing mechanisms, and exhibiting anti-cancer properties. The results showed that the AgNPs-LC had a spherical shape with an average size of 15 nm. The biosynthesized AgNPs-LC showed a symmetrical absorption spectrum with a peak at 458 nm with a diameter of 5-20 nm. AgNPs-LC exhibited significant antibacterial activity against Gram-positive and Gram-negative bacteria and inhibited the biofilm formation (> 50% at sub-MIC) and quorum sensing-mediated virulence factors, such as the production of violacein in C. violaceum (> 80% at sub-MIC), pyocyanin in P. aeruginosa (> 70% at sub-MIC), and prodigiosin in S. marcescens (> 80% at sub-MIC). The exopolysaccharides (EPS) were also found to reduce in the presence of AgNPs-LC. Furthermore, the AgNPs-LC showed anti-cancer and anti-metastasis activity via inhibiting cell migration and invasion of human lung cancer (A-549) cells. Overall, the present study brings out the multifaceted therapeutic capabilities of AgNPs-LC which offer exciting prospects for the development of innovative biomedical and pharmaceutical interventions, making AgNPs-LC a versatile and promising candidate for a wide range of applications in healthcare and medicine. However, further research is essential to fully harness their therapeutic potential.
Collapse
Affiliation(s)
- Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Ha'il, P.O. Box 2440, Ha'il, Saudi Arabia.
| | - Mitesh Patel
- Research and Development Cell, Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Vadodara, 391760, India
| | - Sadaf Jahan
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah, 11952, Saudi Arabia
| | - Abdelmushin Abdelgadir
- Department of Biology, College of Science, University of Ha'il, P.O. Box 2440, Ha'il, Saudi Arabia
| | - Mohammad Jahoor Alam
- Department of Biology, College of Science, University of Ha'il, P.O. Box 2440, Ha'il, Saudi Arabia
| | - Mohammed Merae Alshahrani
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Najran University, 1988, Najran, 61441, Saudi Arabia
| | - Wael Alturaiki
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah, 11952, Saudi Arabia
| | - Manojkumar Sachidanandan
- Department of Oral Radiology, College of Dentistry, University of Ha'il, P.O. Box 2440, Ha'il, Saudi Arabia
| | - Andleeb Khan
- Department of Biosciences, Faculty of Science, Integral University, Lucknow, 22602, India
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan, 45142, Saudi Arabia
| | - Riadh Badraoui
- Department of Biology, College of Science, University of Ha'il, P.O. Box 2440, Ha'il, Saudi Arabia
- Section of Histology-Cytology, Medicine Faculty of Tunis, University of Tunis El Manar, 1017 La Rabta, Tunis, Tunisia
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'il, P.O. Box 2440, Ha'il, Saudi Arabia
| |
Collapse
|
4
|
Vastrad SJ, Saraswathy GR, Dasari JB, Nair G, Madarakhandi A, Augustine D, Sowmya S. A comprehensive transcriptome based meta-analysis to unveil the aggression nexus of oral squamous cell carcinoma. Biochem Biophys Rep 2025; 42:102001. [PMID: 40271514 PMCID: PMC12016861 DOI: 10.1016/j.bbrep.2025.102001] [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: 01/05/2025] [Revised: 03/21/2025] [Accepted: 03/31/2025] [Indexed: 04/25/2025] Open
Abstract
Lymph node metastasis in oral cancer (OC) complicates management due to its aggressive nature and high risk of recurrence, underscoring the need for biomarkers for early detection and targeted therapies. However, the drivers of this aggressive phenotype remain unclear due to the variability in gene expression patterns. To address this, an integrative meta-analysis of six publicly available transcriptomic profiles, categorized by lymph nodal status, is conducted. Key determinants of disease progression are identified through functional characterization and the TopConfects ranking approach of nodal associated differentially expressed genes (DEGs). To explore the critical nexus between lymph node metastasis and OC recurrence, significant metastatic genes were cross-analysed with literature-derived genes exhibiting aberrant methylation patterns in OC recurrence. Their clinical relevance and expression patterns were then validated in an external dataset from the TCGA head and neck cancer cohort. The analysis identified elevated expression of genes involved in extracellular matrix remodelling and immune response, while the expression of genes related to cellular differentiation and barrier functions was reduced, driving the transition to nodal positivity. The highest-ranked gene, MMP1, showed a log-fold change (LFC) of 4.946 (95 % CI: 3.71, 6.18) in nodal-negative samples, which increased to 5.899 (95 % CI: 4.80, 6.99) in nodal-positive samples, indicating consistent elevation across disease stages. In contrast, TMPRSS11B was significantly downregulated, with an LFC of -5.512 (95 % CI: -6.63, -4.38) in nodal-negative samples and -5.898 (95 % CI: -7.15, -4.64) in nodal-positive samples. Furthermore, MEIS1, down-regulated in nodal-positive status, was found to exhibit hypermethylation at CpG sites associated with OC recurrence. This study represents the first transcriptomic meta-analysis to explore the intersection of lymph node metastasis and OC recurrence, identifying MEIS1 as a potential key contributor. These comprehensive insights into disease trajectories offer potential biomarkers and therapeutic targets for future treatment strategies.
Collapse
Affiliation(s)
- Soujanya J. Vastrad
- Department of Pharmacy Practice, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, New BEL Road, M.S.R. Nagar, Bengaluru, India
| | - Ganesan Rajalekshmi Saraswathy
- Department of Pharmacy Practice, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, New BEL Road, M.S.R. Nagar, Bengaluru, India
| | | | - Gouri Nair
- Department of Pharmacology, Faculty of Pharmacy, M. S. Ramaiah University of Applied Sciences, Bangalore, Karnataka, India
| | - Ashok Madarakhandi
- Department of Pharmaceutical Chemistry, KLE College of Pharmacy, (A Constituent Unit of KAHER-Belagavi), 2nd Block, Rajajinagar, Bangalore, India
| | - Dominic Augustine
- Department of Oral Pathology and Microbiology, Faculty of Dental Sciences, M.S. Ramaiah University of Applied Sciences, New BEL Road, M.S.R. Nagar, Bengaluru, India
| | - S.V. Sowmya
- Department of Oral Pathology and Microbiology, Faculty of Dental Sciences, M.S. Ramaiah University of Applied Sciences, New BEL Road, M.S.R. Nagar, Bengaluru, India
| |
Collapse
|
5
|
Zhu K, Bai Y, Lin C, Song G, Chen Y. An energy metabolism-related signature relevant to the tumor immune microenvironment in HNSCC. Discov Oncol 2025; 16:806. [PMID: 40383838 PMCID: PMC12086135 DOI: 10.1007/s12672-025-02652-7] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 05/09/2025] [Indexed: 05/20/2025] Open
Abstract
The importance of energy metabolism in cancer was explored by accumulating studies. Energy metabolism can affect the cellular activities of tumors. However, there is few research exploring the role of energy metabolism in tumor immune microenvironment. In this context, we constructed a novel energy metabolism-related prognostic signature containing 8 genes. The risk score calculated by the signature was analyzed to be an independent value of head and neck squamous cell carcinoma (HNSCC). We further validated the effectiveness and accuracy of our signature in The Cancer Genome Atlas Program (TCGA) cohort and Gene Expression Omnibus (GEO) cohort. Moreover, we also revealed a negative correlation between the risk score and the activity of the immune processes. Finally, we validated the function of Desmoglein 2 protein (DSG2), a risk gene in the signature, in tumor progression and found that knockdown of DSG2 remarkably suppressed the proliferation and migration of HNSCC cells, which further validated our analysis. In conclusion, the energy metabolism-related gene signature we built is a prospective biomarker of HNSCC, which can offer valuable clues for the research and development of immunotherapeutic drugs in HNSCC.
Collapse
Affiliation(s)
- Kaiyu Zhu
- Department of Otolaryngology Head Neck Surgery, The Fourth Hospital of Changsha (Integrated Traditional Chinese and Western Medicine Hospital of Changsha, Changsha Hospital of Hunan Normal University), 200 Jinxing North Road, Changsha, 410219, Hunan, People's Republic of China
- Center for Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, People's Republic of China
| | - Yang Bai
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, People's Republic of China
- Postdoctoral Station of Basic Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, People's Republic of China
| | - Changwei Lin
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, People's Republic of China
| | - Guilin Song
- Department of Otolaryngology Head Neck Surgery, The Fourth Hospital of Changsha (Integrated Traditional Chinese and Western Medicine Hospital of Changsha, Changsha Hospital of Hunan Normal University), 200 Jinxing North Road, Changsha, 410219, Hunan, People's Republic of China
| | - Yifei Chen
- Department of Otolaryngology Head Neck Surgery, The Fourth Hospital of Changsha (Integrated Traditional Chinese and Western Medicine Hospital of Changsha, Changsha Hospital of Hunan Normal University), 200 Jinxing North Road, Changsha, 410219, Hunan, People's Republic of China.
| |
Collapse
|
6
|
Xu C, Wei H, Wang Y, Ma X, Li Q, Bi Y, Li Z, Li Z, Zhu S, Tian J, Quan C, Qi S. The role of adherent-to-suspension transition factors in clear cell renal cell carcinoma progression: a comprehensive analysis. Sci Rep 2025; 15:13618. [PMID: 40253446 PMCID: PMC12009395 DOI: 10.1038/s41598-025-96807-9] [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: 07/17/2024] [Accepted: 03/31/2025] [Indexed: 04/21/2025] Open
Abstract
Recent studies have identified a biological process called adherent-to-suspension transition (AST) as a key factor in promoting metastasis. The involvement of AST in the progression of clear cell renal cell carcinoma (ccRCC) remains largely unexplored. We comprehensively investigated the overall landscape of all 20 AST factors at the pan-cancer level. The risk stratification method and the prognostic model based on AST factors were established with consensus clustering analysis and LASSO regression algorithm. The potential molecular mechanism of AST was further investigated by multi-omics analysis and cell experiments. We found that AST factors play distinct roles in different cancer types. Consequently, we utilized AST factors to develop a risk stratification method and a prognostic model, which can effectively guide the treatment of ccRCC patients. We proposed that the promotion of AST is facilitated by SPIB through the SAA1-AKT pathway, enhancing the likelihood of ccRCC metastasis, and conducted rigorous analyses using multi-omics data and cellular experiments. Our study presents a novel risk stratification method and prognostic model for ccRCC. And we identified the SPIB-SAA1-AKT pathway as one of the potential mechanisms by which AST factors promote ccRCC metastasis.
Collapse
Affiliation(s)
- Chenglong Xu
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Haotian Wei
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yue Wang
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Xiaocheng Ma
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Qi Li
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yingjie Bi
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zifei Li
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhaochen Li
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Shimiao Zhu
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Jing Tian
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.
| | - Changyi Quan
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.
| | - Shiyong Qi
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.
| |
Collapse
|
7
|
Hirao H, Honda M, Tomita M, Li L, Adawy A, Xue W, Hibi T. Intravital Imaging of Immune Responses in the Cancer Microenvironment. Cancer Med 2025; 14:e70899. [PMID: 40257446 PMCID: PMC12010765 DOI: 10.1002/cam4.70899] [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: 02/08/2025] [Revised: 03/18/2025] [Accepted: 04/09/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND To date, many types of immune cells have been identified, but their precise role in cancer immunity remains unclear. Understanding the immune responses involved in cancer and the cancer microenvironment is becoming increasingly important for elucidating disease mechanisms. In recent years, the application of intravital imaging in cancer research has provided new insights into the mechanisms of cancer-specific immune events, including innate and adaptive immunity. RESULTS In this review, we focus on the emerging role of intravital imaging in cancer research and describe how cancer and immune cells can be observed using intravital imaging in vivo. We also discuss new insights gained by this state-of-the-art technique. CONCLUSIONS Intravital imaging is a relatively new field of research that offers significant advantages, including the ability to directly capture cell-cell interactions, pathophysiology, and immune cell dynamics in the cancer microenvironment in vivo.
Collapse
Affiliation(s)
- Hiroki Hirao
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Masaki Honda
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Masahiro Tomita
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Lianbo Li
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Ahmad Adawy
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Weijie Xue
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Taizo Hibi
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| |
Collapse
|
8
|
Duzgun Z, Korkmaz FD, Akgün E. FDI-6 inhibits VEGF-B expression in metastatic breast cancer: a combined in vitro and in silico study. Mol Divers 2025; 29:1069-1078. [PMID: 38853176 PMCID: PMC11909019 DOI: 10.1007/s11030-024-10891-z] [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: 03/20/2024] [Accepted: 05/09/2024] [Indexed: 06/11/2024]
Abstract
Angiogenesis is the process by which new blood vessels are formed to meet the oxygen and nutrient needs of tissues. This process is vitally important in many physiological and pathological conditions such as tumor growth, metastasis, and chronic inflammation. Although the relationship of FDI-6 compound with FOXM1 protein is well known in the literature, its relationship with angiogenesis is not adequately elucidated. This study investigates the relationship of FDI-6 with angiogenesis and vascular endothelial growth factor B (VEGF-B) protein expression alterations. Furthermore, the study aims to elucidate the in silico interaction of FDI-6 with the VEGFR1 protein, a key player in initiating the angiogenic process, which is activated through its binding with VEGF-B. Our results demonstrate a significant effect of FDI-6 on cell viability. Specifically, we determined that the IC50 value of FDI-6 in HUVEC cells after 24 h of treatment is 24.2 μM, and in MDA-MB-231 cells after 24 h of application, it is 10.8 μM. These findings suggest that the cytotoxic effect of FDI-6 varies depending on the cell type. In wound healing experiments, FDI-6 significantly suppressed wound closure in MDA-MB-231 cells but did not show a similar effect in HUVEC cells. This finding suggests FDI-6 may have potential cell-type-specific effects. Molecular docking studies reveal that FDI-6 exhibits a stronger interaction with the VEGFR1 protein compared to its inhibitor, a novel interaction not previously reported in the literature. Molecular dynamic simulation results demonstrate a stable interaction between FDI-6 and VEGFR1. This interaction suggests that FDI-6 might modulate mechanisms associated with angiogenesis. Our Western blot analysis results show regulatory effects of FDI-6 on the expression of the VEGF-B protein. We encourage exploration of FDI-6 as a potential therapeutic agent in pathological processes related to angiogenesis. In conclusion, this study provides a detailed examination of the relationship between FDI-6 and both the molecular interactions and protein expressions of VEGF-B. Our findings support FDI-6 as a potential therapeutic agent in pathological processes associated with angiogenesis.
Collapse
Affiliation(s)
- Zekeriya Duzgun
- Department of Medical Biology, Faculty of Medicine, Giresun University, Giresun, Turkey.
| | | | - Egemen Akgün
- Department of Medical Biology, Faculty of Medicine, Giresun University, Giresun, Turkey
| |
Collapse
|
9
|
Roy D, Bhattacharya B, Chakravarti R, Singh P, Arya M, Kundu A, Patil A, Siva B, Mehta S, Kazi TA, Ghosh D. LncRNAs in oncogenic microenvironment: from threat to therapy. Front Cell Dev Biol 2025; 12:1423279. [PMID: 40176927 PMCID: PMC11962222 DOI: 10.3389/fcell.2024.1423279] [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: 04/25/2024] [Accepted: 12/09/2024] [Indexed: 04/05/2025] Open
Abstract
LncRNAs are RNA molecules of more than 200 nucleotides in length and participate in cellular metabolism and cellular responses through their diverse interactomedespite having no protein-coding capabilities. Such significant interactions also implicate the presence of lncRNAs in complex pathobiological pathways of various diseases, affecting cellular survival by modulating autophagy, inflammation and apoptosis. Proliferating cells harbour a complex microenvironment that mainly stimulate growth-specific activities such as DNA replication, repair, and protein synthesis. They also recognise damages at the macromolecular level, preventing them from reaching the next-generation. LncRNAs have shown significant association with the events occurring towards proliferation, regulating key events in dividing cells, and dysregulation of lncRNA transcriptome affects normal cellular life-cycle, promoting the development of cancer. Furthermore, lncRNAs also demonstrated an association with cancer growth and progression by regulating key pathways governing cell growth, epithelial-mesenchymal transition and metastasis. This makes lncRNAs an attractive target for the treatment of cancer and can also be used as a marker for the diagnosis and prognosis of diseases due to their differential expression in diseased samples. This review delves into the correlation of the lncRNA transcriptome with the fundamental cellular signalling and how this crosstalk shapes the complexity of the oncogenic microhabitat.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Dipanjan Ghosh
- Department of Natural Products, National Institute of Pharmaceutical Education and Research-Kolkata, Kolkata, India
| |
Collapse
|
10
|
Chong Y, Zhou H, Zhang P, Xue L, Du Q, Chong T, Wang Z. Establishing cM0 (i+) stage criteria in localized renal cell carcinoma based on postoperative circulating tumor cells monitoring. BMC Cancer 2025; 25:436. [PMID: 40069681 PMCID: PMC11895218 DOI: 10.1186/s12885-025-13815-8] [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: 09/05/2024] [Accepted: 02/25/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND The diagnostic criteria for cM0 (i+) stage proposed by American Joint Committee on Cancer (AJCC) in renal cell carcinoma (RCC) still remains unclear. The present study aimed to establish and validate the criteria of cM0 (i+) stage based on postoperative circulating tumor cells (CTCs) monitoring in patients with localized renal cell carcinoma (LRCC). MATERIALS AND METHODS This study enrolled 204 patients with LRCC who received partial or radical nephrectomy from January 2015 to November 2021. Cases were randomly divided into test set and validation set. The correlation between clinicopathological features and CTCs counts were analyzed and prognostic variables were determined by Lasso regression. Receiver operating characteristic curve of the prognosis-related CTCs terms were plotted to determine their optimal cut-off value to establish the criteria of cM0 (i+) stage. Its clinical prognostic significance was explored by Kaplan-Meier analysis and Log-rank test. The above analysis was conducted by SPSS26.0 software and R Studio software. P < 0.05 was considered to be statistically significant. RESULTS A total of 204 patients were analyzed in this study.There were no significant differences in variables between the validation and test sets (P>0.05). Total CTCs, mesenchymal CTCs (MCTCs), and CTCs showing a progressive trend were selected as the diagnostic basis for the cM0 (i+) stage through correlation analysis and Lasso regression. The cM0 (i+) stage identified patients meeting the following criteria simultaneously: (1) total CTCs ≥ 6; (2) MCTCs ≥ 1; and (3) a demonstrated trend of progression in either total CTCs or MCTCs. In the validation group, Kaplan-Meier analysis showed that patients with cM0 (i+) stage had significantly shorter progression-free survival than the control group(P<0.05). The results of multivariate Cox regression analysis also showed cM0 (i+) was an independent risk factor for postoperative progression of LRCC patients [12.448 (1.874-82.666) P < 0.05]. Its 1-3 years' prediction discrimination is better than that of UISS score and SSIGN score, which was also verified in the validation set. CONCLUSION The study proposed a diagnostic criterion for M0 (i+) stage in LRCC based on postoperative CTCs monitoring. It was identified as an independent risk factor for postoperative progression and demonstrated potential advantages over the UISS and SSIGN scores in internal validation.
Collapse
Affiliation(s)
- Yue Chong
- Department of Urology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi Wu Road, Xin Cheng district, Xi'an, Shaanxi, 710004, China
| | - Haibin Zhou
- Department of Urology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi Wu Road, Xin Cheng district, Xi'an, Shaanxi, 710004, China
- Department of Urology, First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi, 710077, China
| | - Peng Zhang
- Department of Urology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi Wu Road, Xin Cheng district, Xi'an, Shaanxi, 710004, China
| | - Li Xue
- Department of Urology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi Wu Road, Xin Cheng district, Xi'an, Shaanxi, 710004, China
| | - Qiao Du
- Department of Urology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi Wu Road, Xin Cheng district, Xi'an, Shaanxi, 710004, China
- Baoji People's Hospital, Baoji, 721001, Shaanxi, China
| | - Tie Chong
- Department of Urology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi Wu Road, Xin Cheng district, Xi'an, Shaanxi, 710004, China
| | - Zhenlong Wang
- Department of Urology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi Wu Road, Xin Cheng district, Xi'an, Shaanxi, 710004, China.
| |
Collapse
|
11
|
Mahapatra C, Kishore A, Gawad J, Al-Emam A, Kouzeiha RA, Rusho MA. Review of electrophysiological models to study membrane potential changes in breast cancer cell transformation and tumor progression. Front Physiol 2025; 16:1536165. [PMID: 40110186 PMCID: PMC11920174 DOI: 10.3389/fphys.2025.1536165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 02/14/2025] [Indexed: 03/22/2025] Open
Abstract
The transformation of normal breast cells into cancerous cells is a complex process influenced by both genetic and microenvironmental factors. Recent studies highlight the significant role of membrane potential (Vm) alterations in this transformation. Cancer cells typically exhibit a depolarized resting membrane potential (RMP) compared to normal cells, which correlates with increased cellular activity and more aggressive cancer behavior. These RMP and Vm changes are associated with altered ion channel activity, altered calcium dynamics, mitochondrial dysfunction, modified gap junction communication, and disrupted signaling pathways. Such fluctuations in RMP and Vm influence key processes in cancer progression, including cell proliferation, migration, and invasion. Notably, more aggressive subtypes of breast cancer cells display more frequent and pronounced Vm fluctuations. Understanding the electrical properties of cancer cells provides new insights into their behavior and offers potential therapeutic targets, such as ion channels and Vm regulation. This review synthesizes current research on how various factors modulate membrane potential and proposes an electrophysiological model of breast cancer cells based on experimental and clinical data from the literature. These findings may pave the way for novel pharmacological targets for clinicians, researchers, and pharmacologists in treating breast cancer.
Collapse
Affiliation(s)
| | - Arnaw Kishore
- Microbiology and Immunology, Xavier University School of Medicine, Aruba, Netherlands
| | - Jineetkumar Gawad
- Department of Pharmaceutical Chemistry, VIVA Institute of Pharmacy, Virar, India
| | - Ahmed Al-Emam
- Department of Pathology, College of Medicine, King Khalid University, Asir, Saudi Arabia
| | - Riad Azzam Kouzeiha
- Faculty of Medical Sciences, Lebanese University, Hadath Campus, Beirut, Lebanon
| | - Maher Ali Rusho
- Department of Biomedical Engineering, University of Colorado Boulder, Boulder, CO, United States
| |
Collapse
|
12
|
Dioguardi M, Musella G, Bizzoca ME, Sovereto D, Guerra C, Laterza P, Martella A, Lo Muzio L, Di Domenico M, Cantore S, Ballini A. The Prognostic Role of miR-375 in Head and Neck Squamous Cell Carcinoma: A Systematic Review, Meta-Analysis, and Trial Sequential Analysis. Int J Mol Sci 2025; 26:2183. [PMID: 40076805 PMCID: PMC11900050 DOI: 10.3390/ijms26052183] [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: 01/16/2025] [Revised: 02/21/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Head and Neck Squamous Cell Carcinoma (HNSCC) is a heterogeneous group of malignancies with poor survival outcomes, particularly in advanced stages. Identifying prognostic biomarkers could help improve patient management. miR-375, a small non-coding RNA, has been shown to influence tumor growth and immune responses, making it a candidate biomarker. This study aims to evaluate the role of miR-375 expression in predicting survival outcomes in HNSCC patients. A systematic review and meta-analysis were conducted according to PRISMA guidelines, incorporating data from six studies and the TGCA cohort, encompassing 452 patients. Fixed-effects models were applied to calculate aggregated hazard ratios (HRs) for overall survival (OS). Kaplan-Meier curves were analyzed using the Tierney method, and Trial Sequential Analysis (TSA) was performed to assess statistical power. Low miR-375 expression was associated with poorer OS, with an aggregated HR of 1.23 (95% CI: 1.10-1.37). Subgroup analysis showed consistent trends across oral and laryngeal squamous cell carcinoma. Sensitivity analysis confirmed these findings. TSA revealed that although the number of patients was sufficient, statistical power was insufficient to confirm a predefined risk reduction ratio (RRR) of 49%. Data from the TGCA cohort supported the meta-analysis findings, with an HR for OS of 1.32 (95% CI: 0.96-1.8). Low miR-375 expression is associated with worse survival outcomes in HNSCC patients, indicating its potential as a prognostic biomarker and therapeutic target. However, the retrospective nature of the included studies underscores the need for prospective research to validate these findings.
Collapse
Affiliation(s)
- Mario Dioguardi
- Department of Clinical and Experimental Medicine, University of Foggia, Via Rovelli 50, 71122 Foggia, Italy; (M.D.); (G.M.); (M.E.B.); (D.S.); (C.G.); (P.L.); (L.L.M.); (A.B.)
| | - Gennaro Musella
- Department of Clinical and Experimental Medicine, University of Foggia, Via Rovelli 50, 71122 Foggia, Italy; (M.D.); (G.M.); (M.E.B.); (D.S.); (C.G.); (P.L.); (L.L.M.); (A.B.)
| | - Maria Eleonora Bizzoca
- Department of Clinical and Experimental Medicine, University of Foggia, Via Rovelli 50, 71122 Foggia, Italy; (M.D.); (G.M.); (M.E.B.); (D.S.); (C.G.); (P.L.); (L.L.M.); (A.B.)
| | - Diego Sovereto
- Department of Clinical and Experimental Medicine, University of Foggia, Via Rovelli 50, 71122 Foggia, Italy; (M.D.); (G.M.); (M.E.B.); (D.S.); (C.G.); (P.L.); (L.L.M.); (A.B.)
| | - Ciro Guerra
- Department of Clinical and Experimental Medicine, University of Foggia, Via Rovelli 50, 71122 Foggia, Italy; (M.D.); (G.M.); (M.E.B.); (D.S.); (C.G.); (P.L.); (L.L.M.); (A.B.)
| | - Pietro Laterza
- Department of Clinical and Experimental Medicine, University of Foggia, Via Rovelli 50, 71122 Foggia, Italy; (M.D.); (G.M.); (M.E.B.); (D.S.); (C.G.); (P.L.); (L.L.M.); (A.B.)
| | - Angelo Martella
- DataLab, Department of Engineering for Innovation, University of Salento, 73100 Lecce, Italy;
| | - Lorenzo Lo Muzio
- Department of Clinical and Experimental Medicine, University of Foggia, Via Rovelli 50, 71122 Foggia, Italy; (M.D.); (G.M.); (M.E.B.); (D.S.); (C.G.); (P.L.); (L.L.M.); (A.B.)
| | - Marina Di Domenico
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via De Crecchio, 7, 80138 Naples, Italy;
| | - Stefania Cantore
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via De Crecchio, 7, 80138 Naples, Italy;
| | - Andrea Ballini
- Department of Clinical and Experimental Medicine, University of Foggia, Via Rovelli 50, 71122 Foggia, Italy; (M.D.); (G.M.); (M.E.B.); (D.S.); (C.G.); (P.L.); (L.L.M.); (A.B.)
| |
Collapse
|
13
|
Wei CH, Weng CW, Wu CY, Chen HY, Chang YH, Chang GC, Chen JJW. E3 ligase TRIM8 suppresses lung cancer metastasis by targeting MYOF degradation through K48-linked polyubiquitination. Cell Death Dis 2025; 16:88. [PMID: 39934162 PMCID: PMC11814372 DOI: 10.1038/s41419-025-07421-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 01/14/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
Ubiquitination is a posttranslational modification that regulates tumour progression-associated proteins through the ubiquitin‒proteasome system, making E3 ligases potential antitumour targets. Here, we report that TRIM8, a member of the TRIM family and an E3 ligase, can act as a tumour suppressor in non-small cell lung cancer (NSCLC). Both gain- and loss-of-function experiments revealed that TRIM8 inhibits the proliferation, colony formation, migration and invasion of NSCLC cells. Experiments with a xenograft model showed that TRIM8 expression suppresses tumour metastasis in vivo. Moreover, low expression of TRIM8 was associated with poor overall survival in both the Taiwanese and GEO lung cancer cohorts. TRIM8 overexpression in lung cancer cells reduced MYOF expression, and restoring MYOF rescued cell migration in TRIM8-overexpressing cells. TRIM8 targeted MYOF for K48-linked ubiquitination, facilitating proteasome-mediated degradation and subsequently suppressing the extracellular secretion of MMPs. Our results provide new insights into the contribution of TRIM8 to lung cancer progression, suggesting that TRIM8 is a new biomarker and a novel therapeutic target for lung cancer.
Collapse
Affiliation(s)
- Chi-Hsuan Wei
- Graduate Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Chia-Wei Weng
- Graduate Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
- School of Medicine and Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chih-Ying Wu
- Graduate Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
- Department of Pathology and Laboratory Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hsuan-Yu Chen
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan
| | - Ya-Hsuan Chang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Gee-Chen Chang
- Graduate Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
- School of Medicine and Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Division of Pulmonary Medicine, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Jeremy J W Chen
- Graduate Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan.
- Graduate Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan.
| |
Collapse
|
14
|
Zhou Z, Yang L, Fang Y, Xu R, Wang X, Wang Y, Fang Z. Integrative analysis of anoikis-related prognostic signature to evaluate the immune landscape and predict therapeutic response in stomach adenocarcinoma. Sci Rep 2025; 15:4353. [PMID: 39910129 PMCID: PMC11799156 DOI: 10.1038/s41598-025-88882-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 01/31/2025] [Indexed: 02/07/2025] Open
Abstract
Stomach adenocarcinoma (STAD) is the most prevalent gastrointestinal malignancy and seriously threatens the life of the global population. Anoikis, a process of programmed cell death that occurs when cells detach from the extracellular matrix, is closely associated with tumor invasion and metastasis. In this study, we used the TCGA-STAD database to identify the expression patterns and prognostic relevance of anoikis-related genes (ARGs) in STAD. Functional enrichment analysis was used to explore the potential pathway. LASSO and Cox regression were used to construct anoikis-related prognostic signature. The anoikis risk score (ARS) incorporated 7 genes and stratified patients into highand low-risk subgroups by median value splitting. In addition, external validation was performed based on GSE66229, GSE15459, and GSE84437 cohorts. Nomograms were created based on risk characteristics in combination with clinical variants and the performance of the model was validated with time-dependent AUC, calibration curves, and decision curve analysis (DCA). The prognostic signature indicated that the low-risk subgroup had better outcomes and significant correlations with tumor microenvironment, immune landscape, immunotherapy response, and drug sensitivity. In addition, single-cell analysis displayed the cell types, the subcellular localization of prognostic genes, and the cellular interaction to reveal the potential molecular communication mechanism of anoikis resistance. Finally, in vitro experiments confirmed the critical role of CRABP2 in STAD. The results indicated that CRABP2 knockdown inhibited gastric cancer cell proliferation, migration and invasion, and promoted apoptosis. In summary, ARS can serve as a biomarker for predicting survival outcomes in STAD patients, providing new tools for personalized treatment decisions for STAD patients.
Collapse
Affiliation(s)
- Ziyi Zhou
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No.274 Zhijiang Middle Road, Jinan, Shanghai, 200071, China
| | - Lanlan Yang
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No.274 Zhijiang Middle Road, Jinan, Shanghai, 200071, China
| | - Yuan Fang
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No.274 Zhijiang Middle Road, Jinan, Shanghai, 200071, China
| | - Rongzhong Xu
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No.274 Zhijiang Middle Road, Jinan, Shanghai, 200071, China
| | - Xi Wang
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No.274 Zhijiang Middle Road, Jinan, Shanghai, 200071, China
| | - Yuli Wang
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No.274 Zhijiang Middle Road, Jinan, Shanghai, 200071, China.
| | - Zhihong Fang
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No.274 Zhijiang Middle Road, Jinan, Shanghai, 200071, China.
| |
Collapse
|
15
|
Liu Z, Liu Y, Kang X, Li L, Xiang Y. Subcellular Organelle Targeting as a Novel Approach to Combat Tumor Metastasis. Pharmaceutics 2025; 17:198. [PMID: 40006565 PMCID: PMC11859411 DOI: 10.3390/pharmaceutics17020198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/28/2025] [Accepted: 02/02/2025] [Indexed: 02/27/2025] Open
Abstract
Tumor metastasis, the spread of cancer cells from the primary site to distant organs, remains a formidable challenge in oncology. Central to this process is the involvement of subcellular organelles, which undergo significant functional and structural changes during metastasis. Targeting these specific organelles offers a promising avenue for enhanced drug delivery and metastasis therapeutic efficacy. This precision increases the potency and reduces potential off-target effects. Moreover, by understanding the role of each organelle in metastasis, treatments can be designed to disrupt the metastatic process at multiple stages, from cell migration to the establishment of secondary tumors. This review delves deeply into tumor metastasis processes and their connection with subcellular organelles. In order to target these organelles, biomembranes, cell-penetrating peptides, localization signal peptides, aptamers, specific small molecules, and various other strategies have been developed. In this review, we will elucidate targeting delivery strategies for each subcellular organelle and look forward to prospects in this domain.
Collapse
Affiliation(s)
- Zefan Liu
- Department of General Surgery, First People‘s Hospital of Shuangliu District (West China Airport Hospital of Sichuan University), Chengdu 610200, China; (Z.L.); (Y.L.)
| | - Yang Liu
- Department of General Surgery, First People‘s Hospital of Shuangliu District (West China Airport Hospital of Sichuan University), Chengdu 610200, China; (Z.L.); (Y.L.)
| | - Xin Kang
- Department of General Surgery, First People‘s Hospital of Shuangliu District (West China Airport Hospital of Sichuan University), Chengdu 610200, China; (Z.L.); (Y.L.)
| | - Lian Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China;
| | - Yucheng Xiang
- School of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| |
Collapse
|
16
|
Zhao K, Sun T, Sun Q, Chen Z, Wang T, Yang J, Li L, Zhu Y, Liu X, Yang D, Lin B, Lu N. Nerve Growth Factor Signaling Promotes Nuclear Translocation of TRAF4 to Enhance Tumor Stemness and Metastatic Dormancy Via C-Jun-mediated IL-8 Autocrine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414437. [PMID: 39716976 PMCID: PMC11831473 DOI: 10.1002/advs.202414437] [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: 11/06/2024] [Revised: 12/03/2024] [Indexed: 12/25/2024]
Abstract
Tumor necrosis factor receptor-associated factor 4 (TRAF4), an E3 ubiquitin ligase, is frequently overexpressed in tumors. Although its cytoplasmic role in tumor progression is well-documented, the precise mechanisms underlying its nuclear localization and functional contributions in tumor cells remain elusive. This study demonstrated a positive correlation between the expression of nuclear TRAF4 and both tumor grades and stemness signatures in human cancer tissues. Notably, reduced nuclear TRAF4 led to decreased stemness properties and metastatic dormancy of tumor cells. Conversely, restoring nuclear TRAF4 in TRAF4-knockout (TRAF4-KO) cells augmented these cellular capabilities. Within the nucleus, the TRAF domain of TRAF4 interacted with c-Jun, thereby stimulating its transcriptional activity. This interaction subsequently led to an enhancement of the promoter activity of interleukin-8 (IL-8), which is identified as a mediator of nuclear TRAF4-induced tumor dormancy. Additionally, activation of AKT signaling by nerve growth factor facilitated TRAF4 phosphorylation at Ser242, enhancing its interaction with 14-3-3θ and promoting its nuclear translocation. Importantly, pharmacological modulation of TRAF4 nuclear translocation is found to suppress tumor tumorigenicity and metastasis in tumor models. This study highlights the critical role of nuclear TRAF4 in regulating tumor stemness and dormancy, positioning it as a potential therapeutic target for metastatic and refractory cancers.
Collapse
Affiliation(s)
- Kai Zhao
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionDepartment of PhysiologySchool of Basic Medicine and Clinical PharmacyChina Pharmaceutical University24 TongjiaxiangNanjing210009China
| | - Tifan Sun
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionDepartment of PhysiologySchool of Basic Medicine and Clinical PharmacyChina Pharmaceutical University24 TongjiaxiangNanjing210009China
| | - Qiruo Sun
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionDepartment of PhysiologySchool of Basic Medicine and Clinical PharmacyChina Pharmaceutical University24 TongjiaxiangNanjing210009China
| | - Zhenzhong Chen
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionDepartment of PhysiologySchool of Basic Medicine and Clinical PharmacyChina Pharmaceutical University24 TongjiaxiangNanjing210009China
| | - Tiepeng Wang
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionDepartment of PhysiologySchool of Basic Medicine and Clinical PharmacyChina Pharmaceutical University24 TongjiaxiangNanjing210009China
- School of PharmacyNanjing University of Chinese Medicine138 Xianlin Rd.Nanjing210023China
| | - Jinming Yang
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionDepartment of PhysiologySchool of Basic Medicine and Clinical PharmacyChina Pharmaceutical University24 TongjiaxiangNanjing210009China
- Department of PharmacyThe Second Hospital of NanjingAffiliated Hospital to Nanjing University of Chinese MedicineNanjing210003China
| | - Lei Li
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029China
| | - Yanan Zhu
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionDepartment of PhysiologySchool of Basic Medicine and Clinical PharmacyChina Pharmaceutical University24 TongjiaxiangNanjing210009China
| | - Xinye Liu
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionDepartment of PhysiologySchool of Basic Medicine and Clinical PharmacyChina Pharmaceutical University24 TongjiaxiangNanjing210009China
| | - Dawei Yang
- Department of PharmacyThe Second Hospital of NanjingAffiliated Hospital to Nanjing University of Chinese MedicineNanjing210003China
| | - Binyan Lin
- School of PharmacyNanjing University of Chinese Medicine138 Xianlin Rd.Nanjing210023China
| | - Na Lu
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionDepartment of PhysiologySchool of Basic Medicine and Clinical PharmacyChina Pharmaceutical University24 TongjiaxiangNanjing210009China
| |
Collapse
|
17
|
Sharma P, Chida K, Wu R, Tung K, Hakamada K, Ishikawa T, Takabe K. VEGFA Gene Expression in Breast Cancer Is Associated With Worse Prognosis, but Better Response to Chemotherapy and Immunotherapy. World J Oncol 2025; 16:120-130. [PMID: 39850522 PMCID: PMC11750749 DOI: 10.14740/wjon1993] [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: 10/23/2025] [Accepted: 01/02/2025] [Indexed: 01/25/2025] Open
Abstract
Background Vascular endothelial growth factor-A (VEGFA) is a key inducer of angiogenesis, responsible for generating new blood vessels in the tumor microenvironment (TME) and facilitating metastasis. Notably, Avastin, which targets VEGFA, failed to demonstrate any significant benefit in clinical trials for breast cancer (BC). This study aimed to investigate the clinical relevance of VEGFA gene expression in BC. Methods A total of 7,336 BC patients across eight independent cohorts: ISPY2 (GSE173839), Sweden Cancerome Analysis Network-Breast (SCAN-B) (GSE96058), Molecular Taxonomy of Breast Cancer International Consortium (METABRIC), GSE25066, GSE163882, GSE34138, GSE20194, and The Cancer Genome Atlas (TCGA), were analyzed. The calculated median VEGFA expression level was used to stratify these cohorts into high and low groups. Results High VEGFA was associated with worse disease-free, disease-specific, and overall survival in the METABRIC cohort, with findings supported by the SCAN-B cohort, which also showed worse overall survival (all P < 0.02). High VEGFA expression was seen in triple-negative breast cancer (TNBC) but not in BC with lymph node metastasis. Additionally, there was a significant correlation between high VEGFA expression and higher silent and non-silent mutations, single-nucleotide variant (SNV) neoantigens, homologous recombination defect, intratumoral heterogeneity, in the TCGA cohort. In the TCGA, METABRIC, and SCAN-B cohorts, high VEGFA BC was also associated with higher cell proliferation: higher Ki67 gene expression, higher Nottingham histological grade, and consistent enrichment of all the Hallmark cell proliferation-related gene sets. Unexpectedly, the angiogenesis gene set was not enriched in any of the cohorts and showed no association with infiltrations of lymphatic or blood vascular endothelial cells besides pericytes. High VEGFA BC had significantly less infiltration of anti-cancer immune cells but higher infiltration of pro-cancer immune cells in TCGA, METABRIC, and SCAN-B cohorts. Interestingly, BC, which had a pathological complete response (pCR) after anthracycline- and taxane-based neoadjuvant therapy, was associated with significantly heightened VEGFA expression in both estrogen receptor (ER)+/human epidermal growth factor receptor 2 (HER2)- and TNBC subtypes in the GSE25066 cohort and after immunotherapy in ER+/ HER2- subtype, but not TNBC in the ISPY2 cohort. Conclusions Our research indicates that high VEGFA BC confers high cell proliferation, reduced immune cell infiltration, and poorer survival, but allows better response to anthracycline- and taxane-based chemotherapy, and immunotherapy.
Collapse
Affiliation(s)
- Pia Sharma
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Kohei Chida
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
- Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan
| | - Rongrong Wu
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo 160-8402, Japan
| | - Kaity Tung
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
- Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY 14203, USA
| | - Kenichi Hakamada
- Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan
| | - Takashi Ishikawa
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo 160-8402, Japan
| | - Kazuaki Takabe
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo 160-8402, Japan
- Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY 14203, USA
- Department of Breast and Thyroid Surgery, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8520, Japan
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| |
Collapse
|
18
|
Miranda I, Jahan N, Shevde LA. The metastatic cascade through the lens of therapeutic inhibition. Cell Rep Med 2025; 6:101872. [PMID: 39706193 PMCID: PMC11866422 DOI: 10.1016/j.xcrm.2024.101872] [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: 06/19/2024] [Revised: 10/21/2024] [Accepted: 11/18/2024] [Indexed: 12/23/2024]
Abstract
Metastasis is a main cause of cancer-related death, and a deeper understanding of the metastatic process will inform more targeted and mechanistic approaches that can abrogate challenges in treatment efficacy and toxicity. Several steps throughout the metastatic cascade, from angiogenesis to secondary tumor formation, offer specific vulnerabilities to therapies that can lead to the decline or cessation of metastatic progression. A deeper understanding of the metastatic cascade also allows combination systemic therapies to be used synergistically. In this review, we describe current treatment modalities in the context of multiple steps of the metastatic cascade. We highlight their mechanisms and present their efficacy across multiple cancers. This work also presents targets within the metastatic cascade in need of more research that can advance the landscape of treatments and lead to the goal of metastatic cancer remission.
Collapse
Affiliation(s)
- Ian Miranda
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nusrat Jahan
- Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lalita A Shevde
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
19
|
Buzaglo GBB, Telles GD, Araújo RB, Junior GDS, Ruberti OM, Ferreira MLV, Derchain SFM, Vechin FC, Conceição MS. The Therapeutic Potential of Physical Exercise in Cancer: The Role of Chemokines. Int J Mol Sci 2024; 25:13740. [PMID: 39769501 PMCID: PMC11678861 DOI: 10.3390/ijms252413740] [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: 07/17/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 01/11/2025] Open
Abstract
The global increase in cancer cases and mortality has been associated with inflammatory processes, in which chemokines play crucial roles. These molecules, a subfamily of cytokines, are essential for the migration, adhesion, interaction, and positioning of immune cells throughout the body. Chemokines primarily originate in response to pathogenic stimuli and inflammatory cytokines. They are expressed by lymphocytes in the bloodstream and are divided into four classes (CC, CXC, XC, and CX3C), playing multifaceted roles in the tumor environment (TME). In the TME, chemokines regulate immune behavior by recruiting cells such as tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs), which promote tumor survival. Additionally, they directly influence tumor behavior, promoting pathological angiogenesis, invasion, and metastasis. On the other hand, chemokines can also induce antitumor responses by mobilizing CD8+ T cells and natural killer (NK) cells to the tumor, reducing pro-inflammatory chemokines and enhancing essential antitumor responses. Given the complex interaction between chemokines, the immune system, angiogenic factors, and metastasis, it becomes evident how important it is to target these pathways in therapeutic interventions to counteract cancer progression. In this context, physical exercise emerges as a promising strategy due to its role modulating the expression of anti-inflammatory chemokines and enhancing the antitumor response. Aerobic and resistance exercises have been associated with a beneficial inflammatory profile in cancer, increased infiltration of CD8+ T cells in the TME, and improvement of intratumoral vasculature. This creates an environment less favorable to tumor growth and supports the circulation of antitumor immune cells and chemokines. Therefore, understanding the impact of exercise on the expression of chemokines can provide valuable insights for therapeutic interventions in cancer treatment and prevention.
Collapse
Affiliation(s)
- Glenda B. B. Buzaglo
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Guilherme D. Telles
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (G.D.T.); (F.C.V.)
| | - Rafaela B. Araújo
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Gilmar D. S. Junior
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Olivia M. Ruberti
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Marina L. V. Ferreira
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Sophie F. M. Derchain
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Campinas, Campinas, Sao Paulo 13083-881, Brazil;
| | - Felipe C. Vechin
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (G.D.T.); (F.C.V.)
| | - Miguel S. Conceição
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| |
Collapse
|
20
|
Obaidur Rab S, Altalbawy FMA, Chandra M, Ariffin IA, Kaur P, Rathore G, Rizaev J, Aloraibi F, Najeeb MA, Abdulhussain MA, Zwamel AH. Targeting the lung tumor microenvironment by phytochemicals and their nanoformulations. Pathol Res Pract 2024; 264:155679. [PMID: 39500198 DOI: 10.1016/j.prp.2024.155679] [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: 10/01/2024] [Revised: 10/15/2024] [Accepted: 10/25/2024] [Indexed: 11/30/2024]
Abstract
Lung malignancies are among the most prevalent and foremost causes of tumor-related deaths. Despite significant advancements in the understanding and management of lung cancer, resistance to traditional treatments remains a significant challenge. Understanding and targeting tumor microenvironment (TME) have attracted interest in the recent decade for eliminating various solid tumors. The lung TME has a crucial position in tumor expansion and therapy failure, driving it an engaging target for novel medicinal interventions. Plant-derived products offer a promising avenue for targeting TME due to their diverse chemical structures and biological activities. However, their clinical use is hindered by insufficient bioavailability and also possible systemic toxicity. The use of nanoparticles as delivery vehicles for natural products can overcome these challenges and enhance their therapeutic efficacy. This review article explores the potential of plant-derived products as medicinal agents for targeting lung TME. We provide an outline of the present knowledge of lung TME and explain the mechanisms by which plant-derived products can modulate key components of this microenvironment. The promising impacts and properties of nanoparticles for the delivery of these derivatives into lung tumors will also be discussed. We also review the preclinical and clinical findings for supporting the usefulness of these agents in targeting lung TME. Additionally, we highlight the challenges and forthcoming trends in the development of plant-derived products as targeted therapies for lung cancer, with a particular focus on combination therapies.
Collapse
Affiliation(s)
- Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia.
| | - Muktesh Chandra
- Department of Bioinformatics, Marwadi University Research Center, Faculty of Engineering and Technology, Marwadi University, Rajkot, Gujarat 360003, India
| | - I A Ariffin
- Management and Science University, Shah Alam, Selangor, Malaysia
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab 140307, India
| | - Gulshan Rathore
- Department of Pharmaceutics, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Jasur Rizaev
- Department of Public health and Healthcare management, Rector, Samarkand State Medical University, 18, Amir Temur Street, Samarkand, Uzbekistan
| | - Farah Aloraibi
- Department of Density, Al-Manara College for Medical Sciences, Maysan, Iraq
| | - Maryam Ali Najeeb
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | | | - Ahmed Hussein Zwamel
- Medical laboratory technique college, the Islamic University, Najaf, Iraq; Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Medical laboratory technique college, the Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
21
|
Wang Y, Nong J, Lu B, Gao Y, Hu M, Chen C, Zhang L, Tan J, Yang X, Lin PP, Hu X, Zhang T. Disseminated tumor cells in bone marrow as predictive classifiers for small cell lung cancer patients. JOURNAL OF THE NATIONAL CANCER CENTER 2024; 4:335-345. [PMID: 39735446 PMCID: PMC11674436 DOI: 10.1016/j.jncc.2024.07.003] [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: 03/26/2024] [Revised: 06/27/2024] [Accepted: 07/20/2024] [Indexed: 12/31/2024] Open
Abstract
Background Small cell lung cancer (SCLC) is a highly aggressive disease characterized by early metastasis. Aneuploid CD31- disseminated tumor cells (DTCs) and CD31+ disseminated tumor endothelial cells (DTECs) residing in the bone marrow are generally considered as the initiators of metastatic process. However, the clinical significance of DTCs and DTECs in SCLC remains poorly understood. The aim of this study is to investigate the clinical implications of diverse subtypes of highly heterogeneous DTCs and DTECs in SCLC patients. Methods Subtraction enrichment and immunostaining-fluorescence in situ hybridization (SE-iFISH) was applied to enrich and perform comprehensive morphologic, karyotypic, and phenotypic characterization of aneuploid DTCs and DTECs in 30 patients. Additionally, co-detection of circulating tumor cells (CTCs) and circulating tumor endothelial cells (CTECs) was conducted on 24 of the enrolled patients. Proof-of-concept of the whole exon sequencings (WES) on precisely selected different subtypes of CTCs or DTCs, longitudinally detected from a representative case with pathologically confirmed bone marrow metastasis, was validated to feasibly reveal genetic mutations in these cells. Results DTCs, DTECs and their subtypes were readily detectable in SCLC patients. Comparative analysis revealed that the number of DTCs and DTECs was significantly higher than that of their corresponding CTCs and CTECs (P < 0.001 for both). Positive detection of disseminated tumor microemboli (DTM) or disseminated tumor endothelial microemboli (DTEM) was associated with inferior survival outcomes (P = 0.046 and P = 0.048). Patients with EpCAM+ DTCs detectable displayed significantly lower disease control rate (DCR) (16.67% vs 73.33%, P = 0.019), reduced median progression-free survival (mPFS) and median overall survival (mOS) compared with those with EpCAM- DTCs (P = 0.028 and P = 0.002, respectively). WES analysis indicated that post-treatment DTCs isolated from bone marrow at the time of disease progression shared more homologous somatic gene mutations with pre-treatment CTCs compared with post-treatment CTCs. Conclusions Our findings suggest that bone marrow sampling and characterization of DTC subtypes provided a valuable tool for predicting treatment response and the prognosis in SCLC. Moreover, DTCs inherit a greater amount of homologous somatic information from pre-treatment CTCs, indicating their potential role in disease progression and treatment resistance.
Collapse
Affiliation(s)
- Ying Wang
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Jingying Nong
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Baohua Lu
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Yuan Gao
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Mingming Hu
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Cen Chen
- The First School of Clinical Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Lina Zhang
- Department of Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Jinjing Tan
- Department of Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Xiaomei Yang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China
- Joint Laboratory for Precision Diagnosis and Treatment Translational Research in Malignant Tumors, Gynecologic Oncology Basic and Clinical Research Laboratory, Capital Medical University, Beijing, China
| | | | - Xingsheng Hu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Tongmei Zhang
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
22
|
Singh P, Jay DG. The Role of eHsp90 in Extracellular Matrix Remodeling, Tumor Invasiveness, and Metastasis. Cancers (Basel) 2024; 16:3873. [PMID: 39594828 PMCID: PMC11592750 DOI: 10.3390/cancers16223873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/01/2024] [Accepted: 11/01/2024] [Indexed: 11/28/2024] Open
Abstract
Identifying proteins that act in tumor invasiveness and metastasis remains a critical unmet need in our search for effective cancer therapy. Hsp90, an abundant intracellular chaperone protein, plays a key role in maintaining cell homeostasis, and its elevated activity is pivotal in cancer progression. Due to the reliance of cancer cells on Hsp90's chaperone function to sustain tumor growth and spread, Hsp90 inhibitors have been the subject of numerous clinical trials over the past two decades. However, these efforts have largely been unsuccessful, primarily due to the cellular toxicity caused by pan-Hsp90 inhibitors at doses required for anticancer efficacy. Therefore, novel approaches to target Hsp90 are necessary. An identified subpopulation of Hsp90 located outside cells (eHsp90) may offer a promising alternative as a therapeutic target against cancer. Studies including our own have shown that eHsp90 is released specifically by cancer cells, and eHsp90 has unique interactors and functions extracellularly to promote tumor invasiveness, the initial step in metastasis. Inhibition of eHsp90 has been shown to suppress metastasis in animal models, indicating its therapeutic potential, although the underlying mechanisms remain incompletely understood. Cancer cells modulate the tumor microenvironment (TME) during the invasion, especially the ECM proteins and the state of the ECM is a strong predictor of invasive and metastatic cancer. Given that most of the known eHsp90 clients are ECM proteins or are proteins involved in ECM modulation, ECM remodelling could be the key mechanism through which eHsp90 enhances invasiveness. This review will focus on ECM modulation by eHsp90 as a driver of cancer invasion and metastasis. We will also discuss the potency of inhibiting eHsp90 in inhibiting invasion and metastatic spread in preclinical models and the using circulating Hsp90 patient samples as a biomarker of cancer invasion and metastasis.
Collapse
Affiliation(s)
- Pragya Singh
- Graduate School of Biomedical Sciences, Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Daniel G. Jay
- Graduate School of Biomedical Sciences, Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| |
Collapse
|
23
|
Biray Avci C, Goker Bagca B, Nikanfar M, Takanlou LS, Takanlou MS, Nourazarian A. Tumor microenvironment and cancer metastasis: molecular mechanisms and therapeutic implications. Front Pharmacol 2024; 15:1442888. [PMID: 39600368 PMCID: PMC11588459 DOI: 10.3389/fphar.2024.1442888] [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: 06/03/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024] Open
Abstract
The tumor microenvironment (TME) plays a crucial role in cancer development and metastasis. This review summarizes the current research on how the TME promotes metastasis through molecular pathways, focusing on key components, such as cancer-associated fibroblasts, immune cells, endothelial cells, cytokines, and the extracellular matrix. Significant findings have highlighted that alterations in cellular communication within the TME enable tumor cells to evade immune surveillance, survive, and invade other tissues. This review highlights the roles of TGF-β and VEGF signaling in promoting angiogenesis and extracellular matrix remodeling, which facilitate metastasis. Additionally, we explored how metabolic reprogramming of tumor and stromal cells, influenced by nutrient availability in the TME, drives cancer progression. This study also evaluated the therapeutic strategies targeting these interactions to disrupt metastasis. By providing a multidisciplinary perspective, this study suggests that understanding the molecular basis of the TME can lead to more effective cancer therapies and identify potential avenues for future research. Future research on the TME should prioritize unraveling the molecular and cellular interactions within this complex environment, which could lead to novel therapeutic strategies and personalized cancer treatments. Moreover, advancements in technologies such as single-cell analysis, spatial transcriptomics, and epigenetic profiling offer promising avenues for identifying new therapeutic targets and improving the efficacy of immunotherapies, particularly in the context of metastasis.
Collapse
Affiliation(s)
- Cigir Biray Avci
- Department of Medical Biology, Faculty of Medicine, Ege University, Izmir, Türkiye
| | - Bakiye Goker Bagca
- Department of Medical Biology, Faculty of Medicine, Adnan Menderes University, Aydin, Türkiye
| | - Masoud Nikanfar
- Department of Neurology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
- Student Research Committee, Khoy University of Medical Sciences, Khoy, Iran
| |
Collapse
|
24
|
J M, Sanji AS, Gurav MJ, Megalamani PH, Vanti G, Kurjogi M, Kaulgud R, Kennedy JF, Chachadi VB. Overexpression of sialyl Lewis a carrying mucin-type glycoprotein in prostate cancer cell line contributes to aggressiveness and metastasis. Int J Biol Macromol 2024; 281:136519. [PMID: 39401629 DOI: 10.1016/j.ijbiomac.2024.136519] [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: 08/01/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 10/20/2024]
Abstract
Metastasis-promoting Lewis and sialyl Lewis antigens expressed on glycoproteins such as mucins are frequently displayed on the surface of prostate cancer cells and could thus be ideal candidates as measures of prostate cancer aggressiveness. The current study describes the altered expression of sialyl Lewisa (sLea) antigen attached to glycoproteins and key glycosyltransferases between normal prostate (RWPE-1) and cancerous cell lines (LNCaP and DU145). Our results suggest that the expression of sLea on different glycoproteins correlates with the aggressiveness of prostate cancer cells, as determined by flow cytometry and fluorescence microscopy. Blotting studies revealed that sLea-bearing glycoproteins, similar to mucins, are predominantly expressed in the more aggressive DU145 cells, followed by LNCaP cells. Immunohistochemistry technique showed a gradient of sLea expression, with low levels in low-grade prostate cancer (stage II/III) and increasing levels in high-grade cancer (stage IV), indicating its potential as a prognostic marker. Additionally, in qRT-PCR analysis significant upregulation of the glycosyltransferases GALNT5 and ST3GAL6 was observed, correlating with the increased sLea expression in LNCaP (3.2- and 14.5-fold) and DU145 (3.3- and 23.75-fold) cells. Our data indicates a correlation between sLea selectin ligand expression and prostate cancer aggressiveness. Furthermore, GALNT5 and ST3GAL6 could serve as benchmarks in PCa malignancy.
Collapse
Affiliation(s)
- Manasa J
- P.G. Department of Studies in Biochemistry, Karnatak University, Dharwad 580 003, India
| | - Ashwini S Sanji
- P.G. Department of Studies in Biochemistry, Karnatak University, Dharwad 580 003, India
| | - Maruti J Gurav
- P.G. Department of Studies in Biochemistry, Karnatak University, Dharwad 580 003, India
| | - Prasanna H Megalamani
- P.G. Department of Studies in Biochemistry, Karnatak University, Dharwad 580 003, India
| | - Gulamnabi Vanti
- Multidisciplinary Research Unit (MRU), Karnataka Institute of Medical Sciences, Hubli 05, India
| | - Mahantesh Kurjogi
- Multidisciplinary Research Unit (MRU), Karnataka Institute of Medical Sciences, Hubli 05, India
| | - Ram Kaulgud
- Multidisciplinary Research Unit (MRU), Karnataka Institute of Medical Sciences, Hubli 05, India
| | - John F Kennedy
- Chembiotech Ltd, Kyrewood House, Tenbury Wells WR15 8FF, UK
| | - Vishwanath B Chachadi
- P.G. Department of Studies in Biochemistry, Karnatak University, Dharwad 580 003, India.
| |
Collapse
|
25
|
He X, Ren E, Dong L, Yuan P, Zhu J, Liu D, Wang J. Contribution of PKS+ Escherichia coli to colon carcinogenesis through the inhibition of exosomal miR-885-5p. Heliyon 2024; 10:e37346. [PMID: 39315148 PMCID: PMC11417213 DOI: 10.1016/j.heliyon.2024.e37346] [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: 07/22/2023] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/25/2024] Open
Abstract
Objectives About 90 % of all colorectal cancer (CRC) fatalities are caused by the metastatic spread of primary tumors, which is closely correlated with patient survival and spreads by circulating tumor cells (CTCs). The epithelial-mesenchymal transition (EMT) that characterizes CTCs is associated with a poor prognosis. Organotropic metastasis is dictated by the transmission of miRNAs by cancer-derived exosomes. The purpose of this research is to examine PKS + E's function. Coli in CRC metastases and exosomal miR-885-5p suppression. Methods A cohort of 100 patients (50 CRC, 50 healthy) underwent colonoscopy screenings from February 2018 to August 2021. Exosomes were isolated using ultracentrifugation, and exosomal miRNA was analyzed using sequencing and qPCR. Results Among the patients, 40 tested positive for E. coli (12 CRC, 23 healthy). Serotyping revealed that 68.57 % harbored the PKS gene. Exosomal miR-885-5p levels were significantly altered in CRC patients with PKS + E. coli. Intriguingly, our findings indicate that exosomes derived from EMT-CRC cells did not affect miR-885-5p synthesis in HUVECs. Moreover, we observed that the levels of miR-885-5p in both exosomes and the total CRC-conditioned medium were comparable upon isolation of exosomes from CRC cells. What's more, an increased expression of miR-558-5p within the tumors, and the group that received exosome treatment, as well as the EMT-HCT116 group, exhibited a higher occurrence of distant metastasis. Conclusion PKS + E. By inhibiting exosomal miR-885-5p, coli is linked to CRC metastases, offering a possible target for therapeutic intervention.
Collapse
Affiliation(s)
- Xiaoming He
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Enbo Ren
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Lujia Dong
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Pengfei Yuan
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Jiaxin Zhu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Dechun Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Jianguang Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| |
Collapse
|
26
|
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: 27] [Impact Index Per Article: 27.0] [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
|
27
|
米 源, 李 旭, 王 聪, 蔺 晨, 赵 志, 颜 晓, 史 平, 王 雷. [miR-2110 Affects the Biological Behaviors of Lung Adenocarcinoma by Regulating CDT1]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:1202-1209. [PMID: 39507968 PMCID: PMC11536258 DOI: 10.12182/20240960505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Indexed: 11/08/2024]
Abstract
Objective To investigate the effect of miR-2110 on the biological behaviors, such as cell proliferation, apoptosis, and metastasis, of lung adenocarcinoma (LUAD) cells by means of cell and animal experiments. Methods Bioinformatics websites, including ENCORI, TargetScan, miRTarBase, and Tarbase, were used to analyze the changes in the expression of miR-2110 in LUAD samples and to predict miR-2110 target. LUAD tissue samples and cells were collected and the changes in the expression of miR-2110 were verified through PCR technology. CCK-8 assay, clonogenic assay, Transwell assay, and flow cytometry were conducted to analyze alterations in the functions of LUAD cells. In addition, 10 BALB/c female nude mice aged 6 to 8 weeks were randomly divided into 2 groups, and the effect of miR-2110 on LUAD was investigated by in vivo experiments. Results miR-2110 was significantly decreased in LUAD tissues and cells compared with the normal lung tissues. miR-2110 overexpression inhibited the proliferation and metastasis of LUAD cells and promoted the apoptosis of tumor cells (P<0.05). Bioinformatics prediction and dual luciferase reporter gene assay results confirmed that miR-2110 could target and bind to CDT1. In addition, overexpression of CDT1 gene reversed the proliferation, metastasis, and apoptosis of miR-2110 compared with the miR-2110 overexpression group (P<0.05). Nude mice in vivo experiments showed that miR-2110 overexpression significantly decreased the expression of Ki67, a tumor proliferation index, and vimentin and MMP9, two metastasis indices, compared with the control group. Conclusion miR-2110 can inhibit proliferation and metastasis of LUAD by targeting CDT1, providing a new rationale for the treatment of LUAD.
Collapse
Affiliation(s)
- 源 米
- 河北医科大学第四医院 急诊科 (石家庄 050017)Department of Emergency, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050017, China
| | - 旭哲 李
- 河北医科大学第四医院 急诊科 (石家庄 050017)Department of Emergency, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050017, China
| | - 聪 王
- 河北医科大学第四医院 急诊科 (石家庄 050017)Department of Emergency, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050017, China
| | - 晨曦 蔺
- 河北医科大学第四医院 急诊科 (石家庄 050017)Department of Emergency, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050017, China
| | - 志超 赵
- 河北医科大学第四医院 急诊科 (石家庄 050017)Department of Emergency, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050017, China
| | - 晓菲 颜
- 河北医科大学第四医院 急诊科 (石家庄 050017)Department of Emergency, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050017, China
| | - 平 史
- 河北医科大学第四医院 急诊科 (石家庄 050017)Department of Emergency, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050017, China
| | - 雷 王
- 河北医科大学第四医院 急诊科 (石家庄 050017)Department of Emergency, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050017, China
| |
Collapse
|
28
|
Shreevatsa B, Hegde S, Narayan P, Dharmashekar C, Jain A, Wani TA, Prabhuswamimath SC, Kollur SP, Shivamallu C. Targeting FAK, VEGF, and MTA1 proteins with Terminalia elliptica: a computational approach for anticancer activity. Front Oncol 2024; 14:1427632. [PMID: 39355129 PMCID: PMC11442428 DOI: 10.3389/fonc.2024.1427632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 08/20/2024] [Indexed: 10/03/2024] Open
Abstract
Cancer remains a significant global health challenge, prompting exploration into alternative treatments such as those derived from natural compounds found in traditional medicine. Recent research has underscored the role of proteins like Focal Adhesion Kinase (FAK), Vascular Endothelial Growth Factor (VEGF), and Metastasis-Associated Protein 1 (MTA1) in driving cancer cell proliferation and survival. Here, we investigated the potential of a single molecule to modulate these key proteins involved in metastasis, offering a promising avenue for cancer therapy. Terminalia elliptica, commonly known as Asna, possesses a diverse range of medicinal properties, including antimicrobial, anti-inflammatory, anticancer, antidiabetic, antiaging, hepatoprotective, antioxidant, and neuroprotective activities. Our study aimed to explore the anticancer potential of Terminalia elliptica by identifying bioactive compounds capable of targeting FAK, VEGF, and MTA1 to impede cancer metastasis. Through in silico analysis, we conducted network analysis using Cytoscape to assess the significance of these bioactive compounds in the inhibition of signaling pathways driving metastasis. The utilization of these bioactives as potential candidates for targeted therapy of VEGF, FAK, and MTA1 regulated pathways was preliminarily assessed by Molecular Docking and MD Simulation. Our findings revealed that phytobioactives namely, Chebulinic Acid of Terminalia elliptica, exhibited notable binding affinity and interaction with FAK, and Chebulagic Acid with VEGF, and MTA1. This discovery holds promise as a novel therapeutic approach for combating cancer, offering potential benefits in cancer treatment and management.
Collapse
Affiliation(s)
- Bhargav Shreevatsa
- Department of Biotechnology and Bioinformatics, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Shrivatsa Hegde
- Department of Biotechnology and Bioinformatics, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Prakruthi Narayan
- Department of Biotechnology and Bioinformatics, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Chandan Dharmashekar
- Department of Biotechnology and Bioinformatics, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Anisha Jain
- Department of Microbiology, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Tanveer A Wani
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Samudyata C Prabhuswamimath
- Department of Biotechnology and Bioinformatics, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Shiva Prasad Kollur
- School of Physical Sciences, Amrita Vishwa Vidyapeetham, Mysuru, Karnataka, India
| | - Chandan Shivamallu
- Department of Biotechnology and Bioinformatics, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| |
Collapse
|
29
|
Kawaguchi Y, Okamoto K, Kataoka Y, Shibata K, Saito H, Shiratori T, Ueda K, Ohshio Y, Hanaoka J. Increasing monocytes after lung cancer surgery triggers the outgrowth of distant metastases, causing recurrence. Cancer Immunol Immunother 2024; 73:212. [PMID: 39235612 PMCID: PMC11377378 DOI: 10.1007/s00262-024-03800-8] [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: 02/13/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024]
Abstract
Patients with lung cancer have a high incidence of tumor recurrence even after curative surgical resection. Some reports indicated that immunosuppressive cells induced by surgical stress could contribute to tumor recurrence after surgery; however, the underlying mechanisms are not fully understood. In this study, we found that increased postoperative blood monocytes served as a risk factor for tumor recurrence in 192 patients with non-small cell lung cancer (NSCLC). We established the lung cancer recurrent mouse model after tumor resection and showed that the surgical stress immediately increased the level of serum monocyte chemoattractant protein-1 (MCP-1), which subsequently increased blood monocytes. These blood monocytes were rapidly recruited into distant micrometastases and became tumor growth-promoting tumor associated macrophages (TAMs). Furthermore, even after the blood MCP-1 and monocytes decreased enough 72 h after tumor resection, TAMs in micrometastases remained rich because the MCP-1 secreted by micrometastases themselves continued to recruit monocytes around the tumor. Consequently, tumor resection triggered the outgrowth of distant metastases via the MCP-1-Monocyte-TAM axis. When we administered the MCP-1 inhibitor to the lung cancer recurrent model mice, blood monocytes decreased after tumor resection, and TAMs in micrometastases also dramatically decreased. Finally, peri- and postoperative treatment with the MCP-1 inhibitor suppressed distant metastases after surgery. Targeting the MCP-1-Monocyte-TAM axis may inhibit surgical stress-induced NSCLC recurrence by attenuating postoperative immunosuppressive monocytes in micrometastases.
Collapse
Affiliation(s)
- Yo Kawaguchi
- Division of General Thoracic Surgery, Department of Surgery, Shiga University of Medical Science, Tsukinowacho, Seta, Otsu City, Shiga, 520-2192, Japan.
| | - Keigo Okamoto
- Division of General Thoracic Surgery, Department of Surgery, Shiga University of Medical Science, Tsukinowacho, Seta, Otsu City, Shiga, 520-2192, Japan
| | - Yoko Kataoka
- Division of General Thoracic Surgery, Department of Surgery, Shiga University of Medical Science, Tsukinowacho, Seta, Otsu City, Shiga, 520-2192, Japan
| | - Kohei Shibata
- Division of General Thoracic Surgery, Department of Surgery, Shiga University of Medical Science, Tsukinowacho, Seta, Otsu City, Shiga, 520-2192, Japan
| | - Hiroki Saito
- Division of General Thoracic Surgery, Department of Surgery, Shiga University of Medical Science, Tsukinowacho, Seta, Otsu City, Shiga, 520-2192, Japan
| | - Takuya Shiratori
- Division of General Thoracic Surgery, Department of Surgery, Shiga University of Medical Science, Tsukinowacho, Seta, Otsu City, Shiga, 520-2192, Japan
| | - Keiko Ueda
- Division of General Thoracic Surgery, Department of Surgery, Shiga University of Medical Science, Tsukinowacho, Seta, Otsu City, Shiga, 520-2192, Japan
| | - Yasuhiko Ohshio
- Division of General Thoracic Surgery, Department of Surgery, Shiga University of Medical Science, Tsukinowacho, Seta, Otsu City, Shiga, 520-2192, Japan
| | - Jun Hanaoka
- Division of General Thoracic Surgery, Department of Surgery, Shiga University of Medical Science, Tsukinowacho, Seta, Otsu City, Shiga, 520-2192, Japan
| |
Collapse
|
30
|
Gu X, Wei S, Lv X. Circulating tumor cells: from new biological insights to clinical practice. Signal Transduct Target Ther 2024; 9:226. [PMID: 39218931 PMCID: PMC11366768 DOI: 10.1038/s41392-024-01938-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 05/31/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
The primary reason for high mortality rates among cancer patients is metastasis, where tumor cells migrate through the bloodstream from the original site to other parts of the body. Recent advancements in technology have significantly enhanced our comprehension of the mechanisms behind the bloodborne spread of circulating tumor cells (CTCs). One critical process, DNA methylation, regulates gene expression and chromosome stability, thus maintaining dynamic equilibrium in the body. Global hypomethylation and locus-specific hypermethylation are examples of changes in DNA methylation patterns that are pivotal to carcinogenesis. This comprehensive review first provides an overview of the various processes that contribute to the formation of CTCs, including epithelial-mesenchymal transition (EMT), immune surveillance, and colonization. We then conduct an in-depth analysis of how modifications in DNA methylation within CTCs impact each of these critical stages during CTC dissemination. Furthermore, we explored potential clinical implications of changes in DNA methylation in CTCs for patients with cancer. By understanding these epigenetic modifications, we can gain insights into the metastatic process and identify new biomarkers for early detection, prognosis, and targeted therapies. This review aims to bridge the gap between basic research and clinical application, highlighting the significance of DNA methylation in the context of cancer metastasis and offering new avenues for improving patient outcomes.
Collapse
Affiliation(s)
- Xuyu Gu
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shiyou Wei
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xin Lv
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
31
|
Datta N, Vp S, Parvathy K, A S S, Maliekal TT. ALDH1A1 as a marker for metastasis initiating cells: A mechanistic insight. Exp Cell Res 2024; 442:114213. [PMID: 39173941 DOI: 10.1016/j.yexcr.2024.114213] [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: 06/04/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 08/24/2024]
Abstract
Since metastasis accounts for the majority of cancer morbidity and mortality, attempts are focused to block metastasis and metastasis initiating cellular programs. It is generally believed that hypoxia, reactive oxygen species (ROS) and the dysregulated redox pathways regulate metastasis. Although induction of epithelial to mesenchymal transition (EMT) can initiate cell motility to different sites other than the primary site, the initiation of a secondary tumor at a distant site depends on self-renewal property of cancer stem cell (CSC) property. That subset of metastatic cells possessing CSC property are referred to as metastasis initiating cells (MICs). Among the different cellular intermediates regulating metastasis in response to hypoxia by inducing EMT and self-renewal property, ALDH1A1 is a critical molecule, which can be used as a marker for MICs in a wide variety of malignancies. The cytosolic ALDHs can irreversibly convert retinal to retinoic acid (RA), which initiates RA signaling, important for self-renewal and EMT. The metastasis permissive tumor microenvironment increases the expression of ALDH1A1, primarily through HIF1α, and leads to metabolic reprograming through OXPHOS regulation. The ALDH1A1 expression and its high activity can reprogram the cancer cells with the transcriptional upregulation of several genes, involved in EMT through RA signaling to manifest hybrid EMT or Hybrid E/M phenotype, which is important for acquiring the characteristics of MICs. Thus, the review on this topic highlights the use of ALDH1A1 as a marker for MICs, and reporters for the marker can be effectively used to trace the population in mouse models, and to screen drugs that target MICs.
Collapse
Affiliation(s)
- Nandini Datta
- Cancer Research, Rajiv Gandhi Centre for Biotechnology (BRIC-RGCB), Thiruvananthapuram, Kerala, 695014, India
| | - Snijesh Vp
- Division of Molecular Medicine, St. John's Research Institute, St John's National Academy of Health Sciences, Bangalore, 560034, India
| | - K Parvathy
- Cancer Research, Rajiv Gandhi Centre for Biotechnology (BRIC-RGCB), Thiruvananthapuram, Kerala, 695014, India
| | - Sneha A S
- Cancer Research, Rajiv Gandhi Centre for Biotechnology (BRIC-RGCB), Thiruvananthapuram, Kerala, 695014, India
| | - Tessy Thomas Maliekal
- Cancer Research, Rajiv Gandhi Centre for Biotechnology (BRIC-RGCB), Thiruvananthapuram, Kerala, 695014, India; Regional Centre for Biotechnology, Faridabad, Haryana 121001, India.
| |
Collapse
|
32
|
Zheng Y, Wang D, Beeghly G, Fischbach C, Shattuck MD, O'Hern CS. Computational modeling of the physical features that influence breast cancer invasion into adipose tissue. APL Bioeng 2024; 8:036104. [PMID: 38966325 PMCID: PMC11223776 DOI: 10.1063/5.0209019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/21/2024] [Indexed: 07/06/2024] Open
Abstract
Breast cancer invasion into adipose tissue strongly influences disease progression and metastasis. The degree of cancer cell invasion into adipose tissue depends on both biochemical signaling and the mechanical properties of cancer cells, adipocytes, and other key components of adipose tissue. We model breast cancer invasion into adipose tissue using discrete element method simulations of active, cohesive spherical particles (cancer cells) invading into confluent packings of deformable polyhedra (adipocytes). We quantify the degree of invasion by calculating the interfacial area At between cancer cells and adipocytes. We determine the long-time value of At vs the activity and strength of the cohesion between cancer cells, as well as the mechanical properties of the adipocytes and extracellular matrix in which adipocytes are embedded. We show that the degree of invasion collapses onto a master curve as a function of the dimensionless energy scale Ec , which grows linearly with the cancer cell velocity persistence time and fluctuations, is inversely proportional to the system pressure, and is offset by the cancer cell cohesive energy. WhenE c > 1 , cancer cells will invade the adipose tissue, whereas forE c < 1 , cancer cells and adipocytes remain de-mixed. We also show that At decreases when the adipocytes are constrained by the ECM by an amount that depends on the spatial heterogeneity of the adipose tissue.
Collapse
Affiliation(s)
| | - Dong Wang
- Department of Mechanical Engineering and Materials Science, Yale University, New Haven, Connecticut 06520, USA
| | - Garrett Beeghly
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Mark D. Shattuck
- Benjamin Levich Institute and Physics Department, City College of New York, New York, New York 10031, USA
| | | |
Collapse
|
33
|
Deng R, Li YY, Bai LL, Zhou L, Wang YS. Case report: A case of Savolitinib in the treatment of MET amplification mutation advanced lung adenocarcinoma with rare bilateral breast metastasis. Front Oncol 2024; 14:1450855. [PMID: 39193383 PMCID: PMC11347308 DOI: 10.3389/fonc.2024.1450855] [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: 06/18/2024] [Accepted: 07/19/2024] [Indexed: 08/29/2024] Open
Abstract
Background The distant metastasis of lung cancer primarily occurs in the bones, liver, brain, and lungs, while the breast is an extremely rare site of metastasis. There is very limited literature on the occurrence of breast metastasis from lung cancer, and metastatic lesions in the breast are prone to being misdiagnosed as primary breast cancer, requiring careful attention and differentiation in the clinical diagnostic and treatment process. Case summary The patient, a 63-year-old female, initially presented with an EGFR exon 21 L858R mutated left lung adenocarcinoma in 2017, treated successfully with surgical resection and subsequent monitoring. The relapse of disease occurred in January 2020. Despite maintaining a prolonged progression-free survival (PFS) with first-generation EGFR-TKI Afatinib, disease progression occurred in 2022 without detectable resistance mutations. Transition to second-generation TKI Furmonertinib resulted in poor control, with rapid progression including unusual bilateral breast metastases that exhibited inflammatory breast cancer-like peau d'orange changes. Standard chemotherapy achieved only short-term stability. Upon detecting a MET amplification mutation, treatment with Savolitinib was initiated. Remarkably, this led to significant clinical and radiographic improvement, notably resolving the peau d'orange appearance and reducing multiple lesions across the body. Conclusion This case underscores the importance of continuous genetic profiling and tailored treatment approaches in managing advanced lung adenocarcinoma, particularly when presenting with rare metastatic sites and complex genetic landscapes. The successful application of Savolitinib following the identification of a MET amplification mutation highlights its potential in overcoming resistance mechanisms in NSCLC, providing a significant therapeutic option for similarly challenging cases.
Collapse
Affiliation(s)
- Rui Deng
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan-ying Li
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Liang-liang Bai
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li Zhou
- Department of Radiotherapy Physics & Technology, West China Hospital, Sichuan University, Chengdu, China
| | - Yong-Sheng Wang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
34
|
Ge X, Du G, Zhou Q, Yan B, Yue G. TNNT1 accelerates migration, invasion and EMT progression in lung cancer cells. Thorac Cancer 2024; 15:1749-1756. [PMID: 38973201 PMCID: PMC11320084 DOI: 10.1111/1759-7714.15400] [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: 05/20/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND Clinically, most patients with lung cancer (LC) die from tumor spread and metastasis. Specific metastasis-related molecules can provide reference for clinical prediction of efficacy, evaluation of prognosis, and search for the best treatment plan. Troponin T1 (TNNT1) is highly expressed in various cancer tissues, which affects malignant behavior of tumor cells and is related to patients' survival and prognosis. However, the role and molecular mechanism of TNNT1 in LC invasion and metastasis have not yet been investigated. METHODS Gene expression profiling interactive analysis (GEPIA) online analysis was used to analyze TNNT1 expression in LC tissues. Quantitative real-time-polymerase chain reaction (qRT-PCR) or western blot were performed to measure TNNT1 or epithelial-to-mesenchymal transition (EMT)-related and Wnt/β-catenin pathway-related protein expression in LC cells. After TNNT1 knockdown, cell scratch healing and transwell assays were introduced to assess cell migration and invasion, respectively. RESULTS TNNT1 expression in LC tissues and cells was increased. TNNT1 knockdown notably impaired LC cell migration, invasion and EMT. TNNT1 knockdown inhibited Wnt/β-catenin pathway of LC cells. Lithium chloride (LiCl) addition partially restored the inhibition of TNNT1 knockdown on migration, invasion, EMT and Wnt/β-catenin of LC cells. CONCLUSION TNNT1 knockdown attenuated LC migration, invasion and EMT, possibly through Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Xiaobin Ge
- Department of Acupuncture‐Moxibustion and TuinaQilu Hospital of Shandong UniversityJinanChina
| | - Guangzhong Du
- Department of Acupuncture‐Moxibustion and TuinaQilu Hospital of Shandong UniversityJinanChina
| | - Qingchen Zhou
- Department of Acupuncture‐Moxibustion and TuinaQilu Hospital of Shandong UniversityJinanChina
| | - Bing Yan
- Ankang Hospital of JinanJinanChina
| | - Gonglei Yue
- Department of Acupuncture‐Moxibustion and TuinaQilu Hospital of Shandong UniversityJinanChina
| |
Collapse
|
35
|
de Sousa C, Eksteen C, Riedemann J, Engelbrecht AM. Highlighting the role of CD44 in cervical cancer progression: immunotherapy's potential in inhibiting metastasis and chemoresistance. Immunol Res 2024; 72:592-604. [PMID: 38816670 PMCID: PMC11347469 DOI: 10.1007/s12026-024-09493-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 05/16/2024] [Indexed: 06/01/2024]
Abstract
Cervical cancer affects thousands of women globally with recurring high-risk HPV infections being at the centre of cervical pathology. Oncological treatment strategies are continually challenged by both chemoresistance and metastasis within patients. Although both work hand-in-hand, targeting their individual mechanisms could prove highly beneficial for treatment outcomes. Such targets include the metastatic-promoting stem cell marker, CD44, which is abundant in cervical cancer cells and is common to both chemoresistance and metastatic mechanisms. Seeing that many existing advanced-stage cervical cancer treatment regimes, such as platinum-based chemotherapy regimens, remain limited and are rarely curative, alternative treatment options within the field of immunology are being considered. The use of immune checkpoint inhibition therapy, which targets immune checkpoints, CTLA-4 and PD-1/PD-L1, has shown promise as an alternate standard of care for patients suffering from advanced-stage cervical cancer. Therefore, this review aims to assess whether immune checkpoint inhibition can mitigate the pathological effects of CD44-induced EMT, metastasis, and chemoresistance in cervical cancer patients.
Collapse
Affiliation(s)
- Cayleigh de Sousa
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Mike de Vries Building, C/o Merriman and Bosman Street, Stellenbosch, 7600, South Africa
| | - Carla Eksteen
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Mike de Vries Building, C/o Merriman and Bosman Street, Stellenbosch, 7600, South Africa
| | | | - Anna-Mart Engelbrecht
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Mike de Vries Building, C/o Merriman and Bosman Street, Stellenbosch, 7600, South Africa.
| |
Collapse
|
36
|
Fu Z, Wang MW, Liu YH, Jiao Y. Impact of immunotherapy on liver metastasis. World J Gastrointest Surg 2024; 16:1969-1972. [PMID: 39087120 PMCID: PMC11287679 DOI: 10.4240/wjgs.v16.i7.1969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 05/09/2024] [Accepted: 05/28/2024] [Indexed: 07/22/2024] Open
Abstract
This editorial discusses the article "Analysis of the impact of immunotherapy efficacy and safety in patients with gastric cancer and liver metastasis" published in the latest edition of the World Journal of Gastrointestinal Surgery. Immunotherapy has achieved outstanding success in tumor treatment. However, the presence of liver metastasis (LM) restrains the efficacy of immunotherapy in various tumors, including lung cancer, colorectal cancer, renal cell carcinoma, melanoma, and gastric cancer. A decrease in CD8+ T cells and nature killer cells, along with an increase in macrophages and regulatory T cells, was observed in the microenvironment of LM, leading to immunotherapy resistance. More studies are necessary to determine the best strategy for enhancing the effectiveness of immunotherapy in patients with LM.
Collapse
Affiliation(s)
- Zhuo Fu
- Medical College, Inner Mongolia Minzu University, Tongliao 028000, Inner Mongolia Autonomous Region, China
| | - Ming-Wei Wang
- Ministry of Health Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun 130000, Jilin Province, China
| | - Ya-Hui Liu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Yan Jiao
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| |
Collapse
|
37
|
Peng R, Ma X, Jiang Z, Duan Y, Lv S, Jing W. Integrative analysis of Anoikis-related genes reveals that FASN is a novel prognostic biomarker and promotes the malignancy of bladder cancer via Wnt/β-catenin pathway. Heliyon 2024; 10:e34029. [PMID: 39071712 PMCID: PMC11283158 DOI: 10.1016/j.heliyon.2024.e34029] [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: 04/07/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/30/2024] Open
Abstract
Bladder cancer (BC) exhibits diversity in clinical outcomes and is characterized by heterogeneity. Anoikis, a form of programmed cell death, plays a crucial role in facilitating tumor invasion and metastasis. This study comprehensively investigated the genetic landscape of BC progression, identifying 300 differentially expressed Anoikis-related genes (DE-ARGs) through in-depth analysis of the GSE13507 datasets. Functional enrichment analysis revealed associations with diverse diseases and biological processes. Employing machine learning algorithms, a logistic regression model based on nine marker genes demonstrated superior accuracy in distinguishing BC from normal samples. Validation in TCGA datasets highlighted the prognostic significance of LRP1, FASN, and SIRT6, suggesting their potential as cancer biomarkers. Particularly, FASN emerged as an independent prognostic indicator, regulating BC cell proliferation and metastasis through the Wnt/β-catenin pathway. The study provides crucial insights into altered genetic landscapes and potential therapeutic strategies for BC, emphasizing the significance of FASN in BC prognosis and progression.
Collapse
Affiliation(s)
- Ruoyu Peng
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan, Zhengzhou, 450000, China
| | - Xiaohan Ma
- Department of Laboratory Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of Pregnancy, Zhengzhou, 450000, China
| | - Zhiyun Jiang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan, Zhengzhou, 450000, China
| | - Yu Duan
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan, Zhengzhou, 450000, China
| | - Shaogang Lv
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan, Zhengzhou, 450000, China
| | - Wei Jing
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan, Zhengzhou, 450000, China
| |
Collapse
|
38
|
Chu X, Tian W, Ning J, Xiao G, Zhou Y, Wang Z, Zhai Z, Tanzhu G, Yang J, Zhou R. Cancer stem cells: advances in knowledge and implications for cancer therapy. Signal Transduct Target Ther 2024; 9:170. [PMID: 38965243 PMCID: PMC11224386 DOI: 10.1038/s41392-024-01851-y] [Citation(s) in RCA: 84] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 03/27/2024] [Accepted: 04/28/2024] [Indexed: 07/06/2024] Open
Abstract
Cancer stem cells (CSCs), a small subset of cells in tumors that are characterized by self-renewal and continuous proliferation, lead to tumorigenesis, metastasis, and maintain tumor heterogeneity. Cancer continues to be a significant global disease burden. In the past, surgery, radiotherapy, and chemotherapy were the main cancer treatments. The technology of cancer treatments continues to develop and advance, and the emergence of targeted therapy, and immunotherapy provides more options for patients to a certain extent. However, the limitations of efficacy and treatment resistance are still inevitable. Our review begins with a brief introduction of the historical discoveries, original hypotheses, and pathways that regulate CSCs, such as WNT/β-Catenin, hedgehog, Notch, NF-κB, JAK/STAT, TGF-β, PI3K/AKT, PPAR pathway, and their crosstalk. We focus on the role of CSCs in various therapeutic outcomes and resistance, including how the treatments affect the content of CSCs and the alteration of related molecules, CSCs-mediated therapeutic resistance, and the clinical value of targeting CSCs in patients with refractory, progressed or advanced tumors. In summary, CSCs affect therapeutic efficacy, and the treatment method of targeting CSCs is still difficult to determine. Clarifying regulatory mechanisms and targeting biomarkers of CSCs is currently the mainstream idea.
Collapse
Affiliation(s)
- Xianjing Chu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Wentao Tian
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jiaoyang Ning
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Gang Xiao
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yunqi Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ziqi Wang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhuofan Zhai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Guilong Tanzhu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Jie Yang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Rongrong Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China.
| |
Collapse
|
39
|
Fofana SI, Oraby T, Cobos E, Tripathi MK. Unraveling the Influence of Social, Economic, and Demographic Factors in Texas on Breast Cancer Survival. RESEARCH SQUARE 2024:rs.3.rs-4535192. [PMID: 38978582 PMCID: PMC11230498 DOI: 10.21203/rs.3.rs-4535192/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Background According to the Centers for Disease Control (CDC), breast cancer is the second most common cancer among women in the United States. Affected people are financially challenged due to the high out-of-pocket cost of breast cancer treatment, as it is the most expensive treatment. Using a 16-year cohort study of breast cancer survival data in Texas, we investigate the factors that might explain why some breast cancer patients live longer than others. Methods Performing a survival analysis consisting of the log-rank test, a survival time regression, and Cox proportional hazards regression, we explore the breast cancer survivors' specific attributes to identify the main determinants of survival time. Results Analyses show that the factors: stage, grade, primary site of the cancer, number of cancers each patient has, histology of the cancer, age, race, and income are among the main variables that enlighten why some breast cancer survivors live much longer than others. For instance, compared to White non-Hispanics, Black non-Hispanics have a shorter length of survival with a hazard ratio of (1.282). The best prognostic for White non-Hispanics, Hispanics (all races), and Black non-Hispanics is a woman aged between 40 to 49 years old, diagnosed with localized stage and grade one with Axillary tail of breast as a primary site with only one cancer and with a household income of 75,000.00 and over. Conclusion Policymakers should promote early diagnosis and screening and better assist the older and the poor to improve the survival time for breast cancer patients.
Collapse
|
40
|
Wang X, Bai L, Kong L, Guo Z. Advances in circulating tumor cells for early detection, prognosis and metastasis reduction in lung cancer. Front Oncol 2024; 14:1411731. [PMID: 38974237 PMCID: PMC11224453 DOI: 10.3389/fonc.2024.1411731] [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: 04/03/2024] [Accepted: 06/07/2024] [Indexed: 07/09/2024] Open
Abstract
Globally, lung cancer stands as the leading type of cancer in terms of incidence and is the major source of mortality attributed to cancer. We have outlined the molecular biomarkers for lung cancer that are available clinically. Circulating tumor cells (CTCs) spread from the original location, circulate in the bloodstream, extravasate, and metastasize, forming secondary tumors by invading and establishing a favorable environment. CTC analysis is considered a common liquid biopsy method for lung cancer. We have enumerated both in vivo and ex vivo techniques for CTC separation and enrichment, examined the advantages and limitations of these methods, and also discussed the detection of CTCs in other bodily fluids. We have evaluated the value of CTCs, as well as CTCs in conjunction with other biomarkers, for their utility in the early detection and prognostic assessment of patients with lung cancer. CTCs engage with diverse cells of the metastatic process, interfering with the interaction between CTCs and various cells in metastasis, potentially halting metastasis and enhancing patient prognosis.
Collapse
Affiliation(s)
- Xiaochen Wang
- Department of Pathology and Pathophysiology, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
- Department of Pathology, Cancer Hospital Affiliated to Inner Mongolia Medical University / Peking University Cancer Hospital Inner Mongolia Hospital, Hohhot, Inner Mongolia, China
| | - Lu Bai
- Department of Pathology and Pathophysiology, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
- Department of Pathology, Cancer Hospital Affiliated to Inner Mongolia Medical University / Peking University Cancer Hospital Inner Mongolia Hospital, Hohhot, Inner Mongolia, China
| | - Linghui Kong
- Department of Pathology, Cancer Hospital Affiliated to Inner Mongolia Medical University / Peking University Cancer Hospital Inner Mongolia Hospital, Hohhot, Inner Mongolia, China
| | - Zhijuan Guo
- Department of Pathology, Cancer Hospital Affiliated to Inner Mongolia Medical University / Peking University Cancer Hospital Inner Mongolia Hospital, Hohhot, Inner Mongolia, China
| |
Collapse
|
41
|
Deng Z, Xu M, Ding Z, Kong J, Liu J, Zhang Z, Cao P. ID2 promotes tumor progression and metastasis in thyroid cancer. Endocrine 2024; 84:1051-1063. [PMID: 38195969 PMCID: PMC11208273 DOI: 10.1007/s12020-023-03674-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/20/2023] [Indexed: 01/11/2024]
Abstract
BACKGROUND Inhibitor of DNA Binding 2 (ID2) plays a crucial role in tumor cell proliferation, invasion, metastasis, and stemness. Aberrant ID2 expression is associated with poor prognosis in various cancers. However, the specific function of ID2 in thyroid cancer remain unclear. METHOD The TCGA database were utilized to explore the clinical relevance of ID2 in cancer. GO, KEGG, and TIMER were employed to predict the potential roles of ID2 in cancer. Functional analysis, including CCK-8, colony formation, transwell, wound healing, and sphere formation experiments, were conducted to determine the biological functions of ID2 in human cancers. Western blot (WB), RT-qPCR, and immunohistochemical (IHC) analyses were used to investigate the relationship between ID2 and downstream targets. RESULTS Our study revealed significant overexpression of ID2 in various malignant tumor cells. Knocking ID2 significantly inhibited cancer cell proliferation and invasion, while overexpressing ID2 enhanced these capabilities. Additionally, ID2 mediates resistance of cancer cells to protein kinase B (or Akt) inhibitions. Further WB and IHC experiments indicated that ID2 promotes the phosphorylation activation of phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway, thereby upregulating the expression of downstream proliferation, epithelial-mesenchymal transition (EMT), and stemness-related markers. CONCLUSION We found that ID2 significantly promotes thyroid cancer cell proliferation, migration, EMT, and stemness through the PI3K/Akt pathway. Moreover, ID2 plays a crucial role in regulating cancer immune responses. It may serve as a potential biomarker for enhancing the efficacy of chemotherapy, targeted therapy, and immunotherapy against cancer.
Collapse
Affiliation(s)
- Zhongming Deng
- Department of General Surgery, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
| | - Min Xu
- Department of Anesthesiology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
| | - Zhenghua Ding
- Department of General Surgery, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
| | - Jianqiao Kong
- Department of General Surgery, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
| | - Juanjuan Liu
- Department of Anesthesiology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
| | - Zelin Zhang
- Department of Oncology Department, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China.
| | - Ping Cao
- Department of Oncology Department, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China.
| |
Collapse
|
42
|
Tran HCM, Mbemba E, Mourot N, Faltas B, Rousseau A, Lefkou E, Sabbah M, van Dreden P, Gerotziafas G. The procoagulant signature of cancer cells drives fibrin network formation in tumor microenvironment and impacts its quality. Implications in cancer cell migration and the resistance to anticancer agents. Thromb Res 2024; 238:172-183. [PMID: 38723522 DOI: 10.1016/j.thromres.2024.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/02/2024] [Accepted: 04/15/2024] [Indexed: 05/21/2024]
Abstract
INTRODUCTION Cancer cells induce hypercoagulability in the tumoral microenvironment by expressing Tissue Factor (TF). We aimed to study the impact of the procoagulant signature of cancer cells on the quality and structure of fibrin network. We also studied the impact of fibrin clot shield (FCS) on the efficiency of anticancer agents and the migration of cancer cells. MATERIALS AND METHODS Pancreatic cancer cells BXPC3 and breast cancer cells MDA-MB231 and MCF7, were cultured in the presence of normal Platelet Poor Plasma (PPP), diluted 10 % in conditioning media. Their potential to induce thrombin generation and their fibrinolytic activity were assessed. The structure of fibrin network was analyzed with Scanning Electron Microscopy (SEM). Cancer cells' mobility with fibrin clot and their interactions with fibrin were observed. Cancer cells were treated with paclitaxel (PTX) or 4-hydroxy-tamoxifen (4OHTam) in the presence or absence of FCS. RESULTS Cancer cells, in presence of PPP, induced fibrin network formation. High TF-expressing cancer cells (BXPC3 and MDA-MB23 cells), led to dense fibrin network with fine fibers. Low TF expressing cells MCF7 led to thick fibers. Exogenous TF enhanced the density of fibrin network formed by MCF7 cells. Cancer cells through their inherent profibrinolytic potential migrated within the fiber scaffold. The BXPC3 and MCF7 cells moved in clusters whereas the MDA-MB231 cells moved individually within the fibrin network. FCS decreased the efficiency of PTX and 4OHTam on the viability of cancer cells. CONCLUSIONS The procoagulant signature of cancer cells is determinant for the quality and structure of fibrin network in the microenvironment. Original SEM images show the architecture of "bird's nest"-like fibrin network being in touch with the cell membranes and surrounding cancer cells. Fibrin network constructed by triggering thrombin generation by cancer cells, provides a scaffold for cell migration. Fibrin clot shields protect cancer cells against PTX and 4OHTam.
Collapse
Affiliation(s)
- Huong Chi Mai Tran
- Sorbonne University, INSERM UMR_S_938, Saint-Antoine Research Center (CRSA), Team "Cancer Biology and Therapeutics", Group "Cancer - Angiogenesis - Thrombosis", University Institute of Cancerology (UIC), 34 Rue du Crozatier, F-75012 Paris, France; Clinical Research Department, Diagnostica Stago, 125 Avenue Louis Roche, 92230 Gennevilliers, France
| | - Elisabeth Mbemba
- Sorbonne University, INSERM UMR_S_938, Saint-Antoine Research Center (CRSA), Team "Cancer Biology and Therapeutics", Group "Cancer - Angiogenesis - Thrombosis", University Institute of Cancerology (UIC), 34 Rue du Crozatier, F-75012 Paris, France
| | - Noémie Mourot
- Sorbonne University, INSERM UMR_S_938, Saint-Antoine Research Center (CRSA), Team "Cancer Biology and Therapeutics", Group "Cancer - Angiogenesis - Thrombosis", University Institute of Cancerology (UIC), 34 Rue du Crozatier, F-75012 Paris, France
| | - Beshoy Faltas
- Sorbonne University, INSERM UMR_S_938, Saint-Antoine Research Center (CRSA), Team "Cancer Biology and Therapeutics", Group "Cancer - Angiogenesis - Thrombosis", University Institute of Cancerology (UIC), 34 Rue du Crozatier, F-75012 Paris, France
| | - Aurélie Rousseau
- Clinical Research Department, Diagnostica Stago, 125 Avenue Louis Roche, 92230 Gennevilliers, France
| | - Elmina Lefkou
- Sorbonne University, INSERM UMR_S_938, Saint-Antoine Research Center (CRSA), Team "Cancer Biology and Therapeutics", Group "Cancer - Angiogenesis - Thrombosis", University Institute of Cancerology (UIC), 34 Rue du Crozatier, F-75012 Paris, France
| | - Michèle Sabbah
- Sorbonne University, INSERM UMR_S_938, Saint-Antoine Research Center (CRSA), Team "Cancer Biology and Therapeutics", Group "Cancer - Angiogenesis - Thrombosis", University Institute of Cancerology (UIC), 34 Rue du Crozatier, F-75012 Paris, France
| | - Patrick van Dreden
- Sorbonne University, INSERM UMR_S_938, Saint-Antoine Research Center (CRSA), Team "Cancer Biology and Therapeutics", Group "Cancer - Angiogenesis - Thrombosis", University Institute of Cancerology (UIC), 34 Rue du Crozatier, F-75012 Paris, France; Clinical Research Department, Diagnostica Stago, 125 Avenue Louis Roche, 92230 Gennevilliers, France
| | - Grigoris Gerotziafas
- Sorbonne University, INSERM UMR_S_938, Saint-Antoine Research Center (CRSA), Team "Cancer Biology and Therapeutics", Group "Cancer - Angiogenesis - Thrombosis", University Institute of Cancerology (UIC), 34 Rue du Crozatier, F-75012 Paris, France; Thrombosis Center, Tenon - Saint Antoine University Hospital,Hôpitaux Universitaires Est Parisien, Assitance Publique Hôpitaix de Paris (AP-HP), 4 Rue de la Chine, 75020 Paris, France.
| |
Collapse
|
43
|
Rojas-Solé C, Torres-Herrera B, Gelerstein-Claro S, Medina-Pérez D, Gómez-Venegas H, Alzolay-Sepúlveda J, Chichiarelli S, Saso L, Rodrigo R. Cellular Basis of Adjuvant Role of n-3 Polyunsaturated Fatty Acids in Cancer Therapy: Molecular Insights and Therapeutic Potential against Human Melanoma. APPLIED SCIENCES 2024; 14:4548. [DOI: 10.3390/app14114548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Human melanoma is a highly aggressive malignant tumor originating from epidermal melanocytes, characterized by intrinsic resistance to apoptosis and the reprogramming of proliferation and survival pathways during progression, leading to high morbidity and mortality rates. This malignancy displays a marked propensity for metastasis and often exhibits poor responsiveness to conventional therapies. Fatty acids, such as n-3 polyunsaturated fatty acids (PUFAs) docosahexaenoic and eicosapentaenoic acids, exert various physiological effects on melanoma, with increasing evidence highlighting the anti-tumorigenic, anti-inflammatory, and immunomodulatory properties. Additionally, n-3 PUFAs have demonstrated their ability to inhibit cancer metastatic dissemination. In the context of cancer treatment, n-3 PUFAs have been investigated in conjunction with chemotherapy as a potential strategy to mitigate severe chemotherapy-induced side effects, enhance treatment efficacy and improve safety profiles, while also enhancing the responsiveness of cancer cells to chemotherapy. Furthermore, dietary intake of n-3 PUFAs has been associated with numerous health benefits, including a decreased risk and improved prognosis in conditions such as heart disease, autoimmune disorders, depression and mood disorders, among others. However, the specific mechanisms underlying their anti-melanoma effects and outcomes remain controversial, particularly when comparing findings from in vivo or in vitro experimental studies to those from human trials. Thus, the objective of this review is to present data supporting the potential role of n-3 PUFA supplementation as a novel complementary approach in the treatment of malignant cancers such as melanoma.
Collapse
Affiliation(s)
- Catalina Rojas-Solé
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380000, Chile
| | - Benjamín Torres-Herrera
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380000, Chile
| | - Santiago Gelerstein-Claro
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380000, Chile
| | - Diego Medina-Pérez
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380000, Chile
| | - Haziel Gómez-Venegas
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380000, Chile
| | - Javier Alzolay-Sepúlveda
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380000, Chile
| | - Silvia Chichiarelli
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University of Rome, 00185 Rome, Italy
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Faculty of Pharmacy and Medicine Sapienza University, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Ramón Rodrigo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380000, Chile
| |
Collapse
|
44
|
Németh A, Bányai GL, Dobos NK, Kós T, Gaál A, Varga Z, Buzás EI, Khamari D, Dank M, Takács I, Szász AM, Garay T. Extracellular vesicles promote migration despite BRAF inhibitor treatment in malignant melanoma cells. Cell Commun Signal 2024; 22:282. [PMID: 38778340 PMCID: PMC11110207 DOI: 10.1186/s12964-024-01660-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Extracellular vesicles (EVs) constitute a vital component of intercellular communication, exerting significant influence on metastasis formation and drug resistance mechanisms. Malignant melanoma (MM) is one of the deadliest forms of skin cancers, because of its high metastatic potential and often acquired resistance to oncotherapies. The prevalence of BRAF mutations in MM underscores the importance of BRAF-targeted therapies, such as vemurafenib and dabrafenib, alone or in combination with the MEK inhibitor, trametinib. This study aimed to elucidate the involvement of EVs in MM progression and ascertain whether EV-mediated metastasis promotion persists during single agent BRAF (vemurafenib, dabrafenib), or MEK (trametinib) and combined BRAF/MEK (dabrafenib/trametinib) inhibition.Using five pairs of syngeneic melanoma cell lines, we assessed the impact of EVs - isolated from their respective supernatants - on melanoma cell proliferation and migration. Cell viability and spheroid growth assays were employed to evaluate proliferation, while migration was analyzed through mean squared displacement (MSD) and total traveled distance (TTD) measurements derived from video microscopy and single-cell tracking.Our results indicate that while EV treatments had remarkable promoting effect on cell migration, they exerted only a modest effect on cell proliferation and spheroid growth. Notably, EVs demonstrated the ability to mitigate the inhibitory effects of BRAF inhibitors, albeit they were ineffective against a MEK inhibitor and the combination of BRAF/MEK inhibitors. In summary, our findings contribute to the understanding of the intricate role played by EVs in tumor progression, metastasis, and drug resistance in MM.
Collapse
Affiliation(s)
- Afrodité Németh
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Gréta L Bányai
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Nikolett K Dobos
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Tamás Kós
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Anikó Gaál
- Institute of Materials and Environmental Chemistry; Biological Nanochemistry Research Group, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| | - Zoltán Varga
- Institute of Materials and Environmental Chemistry; Biological Nanochemistry Research Group, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| | - Edit I Buzás
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- ELKH-SE Translational Extracellular Vesicle Research Group, Budapest, Hungary
- HCEMM-SE Extracellular Vesicle Research Group, Budapest, Hungary
| | - Delaram Khamari
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Magdolna Dank
- Department of Internal Medicine and Oncology, Division of Oncology, Semmelweis University, Budapest, Hungary
| | - István Takács
- Department of Internal Medicine and Oncology, Division of Oncology, Semmelweis University, Budapest, Hungary
| | - A Marcell Szász
- Department of Internal Medicine and Oncology, Division of Oncology, Semmelweis University, Budapest, Hungary
| | - Tamás Garay
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary.
- Department of Internal Medicine and Oncology, Division of Oncology, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
45
|
Naponelli V, Rocchetti MT, Mangieri D. Apigenin: Molecular Mechanisms and Therapeutic Potential against Cancer Spreading. Int J Mol Sci 2024; 25:5569. [PMID: 38791608 PMCID: PMC11122459 DOI: 10.3390/ijms25105569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
Due to its propensity to metastasize, cancer remains one of the leading causes of death worldwide. Thanks in part to their intrinsic low cytotoxicity, the effects of the flavonoid family in the prevention and treatment of various human cancers, both in vitro and in vivo, have received increasing attention in recent years. It is well documented that Apigenin (4',5,7-trihydroxyflavone), among other flavonoids, is able to modulate key signaling molecules involved in the initiation of cancer cell proliferation, invasion, and metastasis, including JAK/STAT, PI3K/Akt/mTOR, MAPK/ERK, NF-κB, and Wnt/β-catenin pathways, as well as the oncogenic non-coding RNA network. Based on these premises, the aim of this review is to emphasize some of the key events through which Apigenin suppresses cancer proliferation, focusing specifically on its ability to target key molecular pathways involved in angiogenesis, epithelial-to-mesenchymal transition (EMT), maintenance of cancer stem cells (CSCs), cell cycle arrest, and cancer cell death.
Collapse
Affiliation(s)
- Valeria Naponelli
- Department of Medicine and Surgery, University of Parma, Plesso Biotecnologico Integrato, Via Volturno 39, 43126 Parma, Italy
| | - Maria Teresa Rocchetti
- Department of Clinical and Experimental Medicine, University of Foggia, Via Pinto 1, 71122 Foggia, Italy;
| | - Domenica Mangieri
- Department of Clinical and Experimental Medicine, University of Foggia, Via Pinto 1, 71122 Foggia, Italy;
| |
Collapse
|
46
|
Baylie T, Kasaw M, Getinet M, Getie G, Jemal M, Nigatu A, Ahmed H, Bogale M. The role of miRNAs as biomarkers in breast cancer. Front Oncol 2024; 14:1374821. [PMID: 38812786 PMCID: PMC11133523 DOI: 10.3389/fonc.2024.1374821] [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: 01/22/2024] [Accepted: 04/08/2024] [Indexed: 05/31/2024] Open
Abstract
Breast cancer (BC) is the second most common cause of deaths reported in women worldwide, and therefore there is a need to identify BC patients at an early stage as timely diagnosis would help in effective management and appropriate monitoring of patients. This will allow for proper patient monitoring and effective care. However, the absence of a particular biomarker for BC early diagnosis and surveillance makes it difficult to accomplish these objectives. miRNAs have been identified as master regulators of the molecular pathways that are emphasized in various tumors and that lead to the advancement of malignancies. Small, non-coding RNA molecules known as miRNAs target particular mRNAs to control the expression of genes. miRNAs dysregulation has been linked to the start and development of a number of human malignancies, including BC, since there is compelling evidence that miRNAs can function as tumor suppressor genes or oncogenes. The current level of knowledge on the role of miRNAs in BC diagnosis, prognosis, and treatment is presented in this review. miRNAs can regulate the tumorigenesis of BC through targeting PI3K pathway and can be used as prognostic or diagnostic biomarkers for BC therapy. Some miRNAs, like miR-9, miR-10b, and miR-17-5p, are becoming known as biomarkers of BC for diagnosis, prognosis, and therapeutic outcome prediction. Other miRNAs, like miR-30c, miR-187, and miR-339-5p, play significant roles in the regulation of hallmark functions of BC, including invasion, metastasis, proliferation, resting death, apoptosis, and genomic instability. Other miRNAs, such as miR-155 and miR-210, are circulating in bodily fluids and are therefore of interest as novel, conveniently accessible, reasonably priced, non-invasive methods for the customized care of patients with BC.
Collapse
Affiliation(s)
- Temesgen Baylie
- Department of Biomedical Science, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Mulugeta Kasaw
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, Bahir Dar University, Bahir Dar, Ethiopia
| | - Mamaru Getinet
- Department of Biomedical Science, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Gedefaw Getie
- Department of Biomedical Science, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Mohammed Jemal
- Department of Biomedical Science, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Amare Nigatu
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, Woldia University, Woldia, Ethiopia
| | - Hassen Ahmed
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, Woldia University, Woldia, Ethiopia
| | - Mihiret Bogale
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, Wollo University, Wollo, Ethiopia
| |
Collapse
|
47
|
Zhang Y, He H, He L, Shi B. IL-6 Accelerates the Proliferation and Metastasis of Pancreatic Cancer Cells via the miR-455-5p/IGF-1R Axis. Cancer Biother Radiopharm 2024; 39:255-263. [PMID: 36595346 DOI: 10.1089/cbr.2022.0045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background: Pancreatic cancer (PaC) is a highly malignant gastrointestinal tumor with invasive and metastatic characteristics. Interleukin-6 (IL-6), a negative prognostic marker, contributes to PaC progression. However, the mechanism of IL-6 in PaC is not yet fully understood. Methods: miR-455-5p levels were first tested by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) in PaC tissues or cells. Subsequently, PaC cell-related functions were identified through CCK-8, Transwell, and Western blotting. Changes in miR-455-5p and IGF-1R expression were confirmed using RT-qPCR and Western blotting. miR-455-5p methylation was assessed by bisulfite sequencing PCR. Results: The authors discovered that miR-455-5p was expressed at low levels in PaC tissues and cells, and miR-455-5p expression was observably reduced by IL-6 in PaC cells. In addition, IL-6 dramatically induces miR-455-5p methylation in PaC cells. Functionally, the data revealed that IL-6 could facilitate the malignant properties of PaC cells, including proliferation, epithelial-mesenchymal transition, and metastasis. The authors found that miR-455-5p could suppress the progression of PaC cells by downregulating IGF-1R in PaC cells. Mechanistically, IL-6 downregulated miR-455-5p and upregulated IGF-1R, and miR-455-5p reduced IGF-1R expression through targeted binding. Conclusions: The authors demonstrated that the miR-455-5p/IGF-1R axis is necessary for the induction of IL-6 in PaC progression. The results here may provide a theoretical basis for the application of the IL-6/miR-455-5p/IGF-1R axis in the clinical therapy of PaC.
Collapse
Affiliation(s)
- Yuting Zhang
- Department of Gynaecology and Obstetrics, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Huan He
- Department of Gastroenterology, School of Medicine, Chongqing University Cancer Hospital, Chongqing University, Chongqing, China
| | - Lanying He
- Department of Gastroenterology, School of Medicine, Chongqing University Cancer Hospital, Chongqing University, Chongqing, China
| | - Bing Shi
- Department of Gastroenterology, School of Medicine, Chongqing University Cancer Hospital, Chongqing University, Chongqing, China
| |
Collapse
|
48
|
Wang J, Yan YL, Yu XY, Pan JY, Liu XL, Hong LL, Wang B. Meroterpenoids from Marine Sponge Hyrtios sp. and Their Anticancer Activity against Human Colorectal Cancer Cells. Mar Drugs 2024; 22:183. [PMID: 38667800 PMCID: PMC11051118 DOI: 10.3390/md22040183] [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: 04/08/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Two new meroterpenoids, hyrtamide A (1) and hyrfarnediol A (2), along with two known ones, 3-farnesyl-4-hydroxybenzoic acid methyl ester (3) and dictyoceratin C (4), were isolated from a South China Sea sponge Hyrtios sp. Their structures were elucidated by NMR and MS data. Compounds 2-4 exhibited weak cytotoxicity against human colorectal cancer cells (HCT-116), showing IC50 values of 41.6, 45.0, and 37.3 μM, respectively. Furthermore, compounds 3 and 4 significantly suppressed the invasion of HCT-116 cells while also downregulating the expression of vascular endothelial growth factor receptor 1 (VEGFR-1) and vimentin proteins, which are key markers associated with angiogenesis and epithelial-mesenchymal transition (EMT). Our findings suggest that compounds 3 and 4 may exert their anti-invasive effects on tumor cells by inhibiting the expression of VEGFR-1 and impeding the process of EMT.
Collapse
Affiliation(s)
- Jie Wang
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China; (Y.-L.Y.); (X.-Y.Y.); (J.-Y.P.); (X.-L.L.)
| | - Yue-Lu Yan
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China; (Y.-L.Y.); (X.-Y.Y.); (J.-Y.P.); (X.-L.L.)
| | - Xin-Yi Yu
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China; (Y.-L.Y.); (X.-Y.Y.); (J.-Y.P.); (X.-L.L.)
| | - Jia-Yan Pan
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China; (Y.-L.Y.); (X.-Y.Y.); (J.-Y.P.); (X.-L.L.)
| | - Xin-Lian Liu
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China; (Y.-L.Y.); (X.-Y.Y.); (J.-Y.P.); (X.-L.L.)
| | - Li-Li Hong
- Research Center for Marine Drugs, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Bin Wang
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China; (Y.-L.Y.); (X.-Y.Y.); (J.-Y.P.); (X.-L.L.)
| |
Collapse
|
49
|
Ozmen E, Demir TD, Ozcan G. Cancer-associated fibroblasts: protagonists of the tumor microenvironment in gastric cancer. Front Mol Biosci 2024; 11:1340124. [PMID: 38562556 PMCID: PMC10982390 DOI: 10.3389/fmolb.2024.1340124] [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: 11/17/2023] [Accepted: 01/31/2024] [Indexed: 04/04/2024] Open
Abstract
Enhanced knowledge of the interaction of cancer cells with their environment elucidated the critical role of tumor microenvironment in tumor progression and chemoresistance. Cancer-associated fibroblasts act as the protagonists of the tumor microenvironment, fostering the metastasis, stemness, and chemoresistance of cancer cells and attenuating the anti-cancer immune responses. Gastric cancer is one of the most aggressive cancers in the clinic, refractory to anti-cancer therapies. Growing evidence indicates that cancer-associated fibroblasts are the most prominent risk factors for a poor tumor immune microenvironment and dismal prognosis in gastric cancer. Therefore, targeting cancer-associated fibroblasts may be central to surpassing resistance to conventional chemotherapeutics, molecular-targeted agents, and immunotherapies, improving survival in gastric cancer. However, the heterogeneity in cancer-associated fibroblasts may complicate the development of cancer-associated fibroblast targeting approaches. Although single-cell sequencing studies started dissecting the heterogeneity of cancer-associated fibroblasts, the research community should still answer these questions: "What makes a cancer-associated fibroblast protumorigenic?"; "How do the intracellular signaling and the secretome of different cancer-associated fibroblast subpopulations differ from each other?"; and "Which cancer-associated fibroblast subtypes predominate specific cancer types?". Unveiling these questions can pave the way for discovering efficient cancer-associated fibroblast targeting strategies. Here, we review current knowledge and perspectives on these questions, focusing on how CAFs induce aggressiveness and therapy resistance in gastric cancer. We also review potential therapeutic approaches to prevent the development and activation of cancer-associated fibroblasts via inhibition of CAF inducers and CAF markers in cancer.
Collapse
Affiliation(s)
- Ece Ozmen
- Koç University Graduate School of Health Sciences, Istanbul, Türkiye
| | - Tevriz Dilan Demir
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
| | - Gulnihal Ozcan
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
- Department of Medical Pharmacology, Koç University School of Medicine, Istanbul, Türkiye
| |
Collapse
|
50
|
Guo B, Zheng Q, Jiang Y, Zhan Y, Huang W, Chen Z. Long non-coding RNAFOXD1-AS1 modulated CTCs epithelial-mesenchymal transition and immune escape in hepatocellular carcinoma in vitro by sponging miR-615-3p. Cancer Rep (Hoboken) 2024; 7:e2050. [PMID: 38517478 PMCID: PMC10959247 DOI: 10.1002/cnr2.2050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 02/26/2024] [Accepted: 03/05/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is widely recognized as a globally prevalent malignancy. Immunotherapy is a promising therapy for HCC patients. Increasing evidence suggests that lncRNAs are involved in HCC progression and immunotherapy. AIM The study reveals the mechanistic role of long non-coding RNA (lncRNA) FOXD1-AS1 in regulating migration, invasion, circulating tumor cells (CTCs), epithelial-mesenchymal transition (EMT), and immune escape in HCC in vitro. METHODS This study employed real-time PCR (RT-qPCR) to measure FOXD1-AS1, miR-615-3p, and programmed death-ligand 1 (PD-L1). The interactions of FOXD1-AS1, miR-615-3p, and PD-L1 were validated via dual-luciferase reporter gene and ribonucleoprotein immunoprecipitation (RIP) assay. In vivo experimentation involves BALB/c mice and BALB/c nude mice to investigate the impact of HCC metastasis. RESULTS The upregulation of lncRNA FOXD1-AS1 in malignant tissues significantly correlates with poor prognosis. The investigation was implemented on the impact of lncRNA FOXD1-AS1 on the migratory, invasive, and EMT of HCC cells. It has been observed that the lncRNA FOXD1-AS1 significantly influences the generation and metastasis of MCTC in vivo analysis. In mechanistic analysis, lncRNA FOXD1-AS1 enhanced immune escape in HCC via upregulation of PD-L1, which acted as a ceRNA by sequestering miR-615-3p. Additionally, lncRNA FOXD1-AS1 was found to modulate the EMT of CTCs through the activation of the PI3K/AKT pathway. CONCLUSION This study presents compelling evidence supporting the role of lncRNA FOXD1-AS1 as a miRNA sponge that sequesters miR-655-3p and protects PD-L1 from suppression.
Collapse
Affiliation(s)
- Bao‐ling Guo
- Department of OncologyLongyan First Affiliated Hospital of Fujian Medical UniversityLongyanFujianPeople's Republic of China
| | - Qiu‐xiang Zheng
- Department of OncologyLongyan First Affiliated Hospital of Fujian Medical UniversityLongyanFujianPeople's Republic of China
| | - Yun‐shan Jiang
- Department of OncologyLongyan First Affiliated Hospital of Fujian Medical UniversityLongyanFujianPeople's Republic of China
| | - Ying Zhan
- Department of OncologyLongyan First Affiliated Hospital of Fujian Medical UniversityLongyanFujianPeople's Republic of China
| | - Wen‐jin Huang
- Department of OncologyLongyan First Affiliated Hospital of Fujian Medical UniversityLongyanFujianPeople's Republic of China
| | - Zhi‐yong Chen
- Department of OncologyLongyan First Affiliated Hospital of Fujian Medical UniversityLongyanFujianPeople's Republic of China
| |
Collapse
|