1
|
Bellazzo A, Montico B, Guerrieri R, Colizzi F, Steffan A, Polesel J, Fratta E. Unraveling the role of hypoxia-inducible factors in cutaneous melanoma: from mechanisms to therapeutic opportunities. Cell Commun Signal 2025; 23:177. [PMID: 40205422 PMCID: PMC11984274 DOI: 10.1186/s12964-025-02173-4] [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: 02/14/2025] [Accepted: 03/24/2025] [Indexed: 04/11/2025] Open
Abstract
Hypoxia is a common feature of solid malignancies, including cutaneous melanoma (CM). Hypoxia-inducible factor (HIF)-1α and HIF-2α orchestrate cellular responses to hypoxia and coordinate a transcriptional program that promote several aggressive features in CM, such as angiogenesis, epithelial-mesenchymal transition, metastasis formation, metabolic rewiring, and immune escape. BRAFV600E, which is the most frequent mutation observed in CM patients, usually increases HIF-α signaling not only in hypoxia, but also in normoxic CM cells, enabling HIF-1α and HIF-2α to continuously activate downstream molecular pathways. In this review, we aim to provide a comprehensive overview of the intricate role and regulation of HIF-1α and HIF-2α in CM, with a brief focus on the complex interactions between HIF-α subunits and non-coding RNAs. We also discuss HIF-α-mediated cellular responses in normoxia along with the mechanisms that allow HIF-α subunits to maintain their stability under normal oxygen conditions. Finally, we resume available evidence on potential therapeutic approaches aimed at targeting HIF-1α and/or HIF-2α.
Collapse
Affiliation(s)
- Arianna Bellazzo
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, via Franco Gallini, 2, Aviano, 33081, PN, Italy
| | - Barbara Montico
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, via Franco Gallini, 2, Aviano, 33081, PN, Italy.
| | - Roberto Guerrieri
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, via Franco Gallini, 2, Aviano, 33081, PN, Italy
| | - Francesca Colizzi
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, via Franco Gallini, 2, Aviano, 33081, PN, Italy
| | - Agostino Steffan
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, via Franco Gallini, 2, Aviano, 33081, PN, Italy
| | - Jerry Polesel
- Unit of Cancer Epidemiology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, via Franco Gallini, 2, Aviano, 33081, PN, Italy
| | - Elisabetta Fratta
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, via Franco Gallini, 2, Aviano, 33081, PN, Italy.
| |
Collapse
|
2
|
Ma J, Cai Y, Lu Y, Fang X. Analysis and assessment of ferroptosis-related gene signatures and prognostic risk models in skin cutaneous melanoma. Transl Cancer Res 2025; 14:1857-1873. [PMID: 40224981 PMCID: PMC11985187 DOI: 10.21037/tcr-24-1506] [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: 08/24/2024] [Accepted: 01/14/2025] [Indexed: 04/15/2025]
Abstract
Background The occurrence and development of skin cutaneous melanoma (SKCM) are significantly influenced by ferroptosis, a sort of regulated cell death characterized by iron deposition and lipid peroxidation. Although positive strides have been achieved in the present management of SKCM, it is still unknown exactly how ferroptosis occurs in this condition. We aimed to determine the role of prognostically relevant ferroptosis-related genes (PR-FRGs) in SKCM development and prognosis. Methods The training group was created using combined transcriptomic RNA data acquired from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases. The dataset GSE19234 was acquired from the Gene Expression Omnibus (GEO) database as a validation group. Differentially expressed ferroptosis-related genes (DE-FRGs) were obtained from the training group, of which 103 showed up-regulation and 77 showed down-regulation. Then, 12 PR-FRGs were identified by the protein-protein interaction (PPI) network and Cox regression analysis, and prognostic risk models and nomograms were constructed. The risk model was validated using a validation group, and the prognostic value of the risk model was analyzed. Finally, immunohistochemical data were obtained from the Human Protein Atlas (HPA) website to validate the PR-FRGs. Results Twelve PR-FRGs were identified. A prognostic risk model was built using PR-FRGs, and patients in the training and validation groups were classified as high or low risk based on the risk model. The outcomes demonstrated that the prognosis was better for the low-risk group. Prognostic value analysis showed that the prognostic risk model could accurately predict the patients' overall survival (OS), was superior to clinical traits such as age, gender, and tumor stage in predicting ability, and could be used as an independent predictor. Meanwhile, the nomogram constructed based on PR-FRGs can effectively predict the prognosis of SKCM patients. Finally, PR-FRGs were validated in the HPA database. Conclusions Ferroptosis affects the prognosis of SKCM patients. Prognostic risk model and nomogram constructed based on 12 PR-FRGs demonstrated significant advantages in predicting the prognosis of SKCM patients. This will help in the identification and prognostic prediction of SKCM and in the discovery of new individualized treatment modalities.
Collapse
Affiliation(s)
- Jianchao Ma
- Department of Orthopedics, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yang Cai
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Youqi Lu
- Department of Orthopedics, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Xu Fang
- Department of Orthopedics, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
3
|
Zhang X, Yu X, Shen Q, Jiang X, Zhou Y, Xue Q, Cao G. The role of TMSB15A in gastric cancer progression and its prognostic significance. J Gastrointest Oncol 2025; 16:27-40. [PMID: 40115917 PMCID: PMC11921194 DOI: 10.21037/jgo-2025-64] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 02/16/2025] [Indexed: 03/23/2025] Open
Abstract
Background Human thymosin β15 (TMSB15A) has been found to have protumorigenic effects in various malignant tumors, yet its function in gastric cancer (GC) remains unclear. This study investigated the value and function of TMSB15A in the diagnosis and tumorigenesis of GC, respectively. Methods Expression data for TMSB15A in GC tissues were analyzed using The Cancer Genome Atlas (TCGA). We evaluated the prognostic significance of TMSB15A through Kaplan-Meier survival analysis, time-dependent receiver operating characteristic (ROC) curves, and Cox regression models. Gene set enrichment analysis (GSEA) was performed to identify pathways associated with TMSB15A. In vitro assays assessed the effects of TMSB15A knockdown on GC cell proliferation, migration, and invasion. Results TMSB15A was significantly overexpressed in GC tissues compared to normal tissues (P<0.05). ROC analysis showed high diagnostic accuracy for TMSB15A (area under the curve =0.851, 95% confidence interval: 0.786-0.905, P<0.05). Kaplan-Meier survival analysis revealed that high TMSB15A expression was associated with poor overall survival, disease-specific survival, and progression-free survival. TMSB15A levels were correlated with advanced tumor stages (P<0.05), lymph node metastasis (P<0.01), and perineural invasion (P<0.05). GSEA showed significant enrichment of TMSB15A in inflammatory and oncogenic pathways, including interleukin-6 (IL-6)/Janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3), transforming growth factor β (TGF-β), and Hedgehog. Functional assays demonstrated that TMSB15A knockdown significantly reduced GC cell proliferation, migration, and invasion, suggesting that TMSB15A contributes to GC tumorigenesis and metastasis. Conclusions TMSB15A could serve as a prospective therapeutic target for GC due to its involvement in disease progression and metastasis.
Collapse
Affiliation(s)
- Xiaolei Zhang
- Department of General Surgery, Nantong Tumor Hospital and Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Xiang Yu
- Department of General Surgery, Nantong Tumor Hospital and Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Qicheng Shen
- Department of General Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Xiaohui Jiang
- Department of General Surgery, Nantong Tumor Hospital and Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Yuan Zhou
- Department of General Surgery, Nantong Tumor Hospital and Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Qiu Xue
- Department of General Surgery, Nantong Tumor Hospital and Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Guangxin Cao
- Department of General Surgery, Nantong Tumor Hospital and Affiliated Tumor Hospital of Nantong University, Nantong, China
| |
Collapse
|
4
|
Zhang MG, Gallo RA, Tan CH, Camacho M, Fasih-Ahmad S, Moeyersoms AHM, Sayegh Y, Dubovy SR, Pelaez D, Rong AJ. Single-Cell RNA Profiling of Ocular Adnexal Sebaceous Carcinoma Reveals a Complex Tumor Microenvironment and Identifies New Biomarkers. Am J Ophthalmol 2025; 270:8-18. [PMID: 39393421 PMCID: PMC11735305 DOI: 10.1016/j.ajo.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/23/2024] [Accepted: 10/03/2024] [Indexed: 10/13/2024]
Abstract
PURPOSE Ocular adnexal sebaceous carcinoma (OaSC) is an aggressive malignancy that often necessitates orbital exenteration. Its tumor composition and transcriptional profile remain largely unknown, which poses a significant barrier to medical advances. Here, we report the first in-depth transcriptomic analysis of OaSC at the single-cell resolution and discern mechanisms underlying cancer progression for the discovery of potential globe-sparing immunotherapies, targeted therapies, and biomarkers to guide clinical management. DESIGN Laboratory investigation with a retrospective observational case series. METHODS Single-cell RNA sequencing was performed on six patient specimens: three primary tumors, two tumors with pagetoid spread, and a normal tarsus sample. Cellular components were identified via gene signatures. Molecular pathways underlying tumorigenesis and pagetoid spread were discerned via gene ontology analysis of the differentially expressed genes between specimens. CALML5 immunohistochemistry was performed on an archival cohort of OaSC, squamous cell carcinoma, ocular surface squamous neoplasia (OSSN), and basal cell carcinoma cases. RESULTS Analysis of 29,219 cells from OaSC specimens revealed tumor, immune, and stromal cells. Tumor-infiltrating immune cells include a diversity of cell types, including exhausted T-cell populations. In primary OaSC tumors, mitotic nuclear division and oxidative phosphorylation pathways are upregulated, while lipid biosynthesis and metabolism pathways are downregulated. Epithelial tissue migration pathways are upregulated in tumor cells undergoing pagetoid spread. Single-cell RNA sequencing analyses also revealed that CALML5 is upregulated in OaSC tumor cells. Diffuse nuclear and cytoplasmic CALML5 staining was present in 28 of 28 (100%) OaSC cases. Diffuse nuclear and membranous CALML5 staining was present in 5 of 25 (20%) squamous cell carcinoma and OSSN cases, while diffuse nuclear staining was present in 1 of 12 (8%) basal cell carcinoma cases. CONCLUSIONS This study reveals a complex OaSC tumor microenvironment and confirms that the CALML5 immunohistochemical stain is a sensitive diagnostic marker.
Collapse
Affiliation(s)
- Michelle G Zhang
- From the Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center (M.G.Z., R.A.G., A.H.M., D.P., and A.J.R.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA; Sylvester Comprehensive Cancer Center (M.G.Z., R.A.G., D.P., and A.J.R.), University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ryan A Gallo
- From the Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center (M.G.Z., R.A.G., A.H.M., D.P., and A.J.R.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA; Sylvester Comprehensive Cancer Center (M.G.Z., R.A.G., D.P., and A.J.R.), University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Charissa H Tan
- Department of Ophthalmology (C.H.T., M.C., S.F.A., Y.S., and S.R.D.), Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Matthew Camacho
- Department of Ophthalmology (C.H.T., M.C., S.F.A., Y.S., and S.R.D.), Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Sohaib Fasih-Ahmad
- Department of Ophthalmology (C.H.T., M.C., S.F.A., Y.S., and S.R.D.), Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Acadia H M Moeyersoms
- From the Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center (M.G.Z., R.A.G., A.H.M., D.P., and A.J.R.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA; Sylvester Comprehensive Cancer Center (M.G.Z., R.A.G., D.P., and A.J.R.), University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Yoseph Sayegh
- Department of Ophthalmology (C.H.T., M.C., S.F.A., Y.S., and S.R.D.), Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Sander R Dubovy
- Department of Ophthalmology (C.H.T., M.C., S.F.A., Y.S., and S.R.D.), Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Daniel Pelaez
- From the Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center (M.G.Z., R.A.G., A.H.M., D.P., and A.J.R.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA; Sylvester Comprehensive Cancer Center (M.G.Z., R.A.G., D.P., and A.J.R.), University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Andrew J Rong
- From the Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center (M.G.Z., R.A.G., A.H.M., D.P., and A.J.R.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA; Sylvester Comprehensive Cancer Center (M.G.Z., R.A.G., D.P., and A.J.R.), University of Miami Miller School of Medicine, Miami, Florida, USA; Division of Oculofacial Plastic, Reconstructive, and Orbital Surgery (A.J.R.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA.
| |
Collapse
|
5
|
Myers BL, Brayer KJ, Paez-Beltran LE, Villicana E, Keith MS, Suzuki H, Newville J, Anderson RH, Lo Y, Mertz CM, Kollipara RK, Borromeo MD, Lu QR, Bachoo RM, Johnson JE, Vue TY. Transcription factors ASCL1 and OLIG2 drive glioblastoma initiation and co-regulate tumor cell types and migration. Nat Commun 2024; 15:10363. [PMID: 39609428 PMCID: PMC11605073 DOI: 10.1038/s41467-024-54750-9] [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: 10/30/2023] [Accepted: 11/20/2024] [Indexed: 11/30/2024] Open
Abstract
Glioblastomas (GBMs) are highly aggressive, infiltrative, and heterogeneous brain tumors driven by complex genetic alterations. The basic-helix-loop-helix (bHLH) transcription factors ASCL1 and OLIG2 are dynamically co-expressed in GBMs; however, their combinatorial roles in regulating the plasticity and heterogeneity of GBM cells are unclear. Here, we show that induction of somatic mutations in subventricular zone (SVZ) progenitor cells leads to the dysregulation of ASCL1 and OLIG2, which then function redundantly and are required for brain tumor formation in a mouse model of GBM. Subsequently, the binding of ASCL1 and OLIG2 to each other's loci and to downstream target genes then determines the cell types and degree of migration of tumor cells. Single-cell RNA sequencing (scRNA-seq) reveals that a high level of ASCL1 is key in specifying highly migratory neural stem cell (NSC)/astrocyte-like tumor cell types, which are marked by upregulation of ribosomal protein, oxidative phosphorylation, cancer metastasis, and therapeutic resistance genes.
Collapse
Affiliation(s)
- Bianca L Myers
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Kathryn J Brayer
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
| | - Luis E Paez-Beltran
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Estrella Villicana
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Matthew S Keith
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Hideaki Suzuki
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Jessie Newville
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Rebekka H Anderson
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Yunee Lo
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Conner M Mertz
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Rahul K Kollipara
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mark D Borromeo
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Q Richard Lu
- Department of Pediatrics, Brain Tumor Center, EHCB, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Robert M Bachoo
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jane E Johnson
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tou Yia Vue
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA.
| |
Collapse
|
6
|
Müller C, Oliveira-Ferrer L, Müller V, Schmalfeldt B, Windhorst S. Transcriptome-based identification of key actin-binding proteins associated with high metastatic potential in breast cancer. Front Mol Biosci 2024; 11:1440276. [PMID: 39281318 PMCID: PMC11392851 DOI: 10.3389/fmolb.2024.1440276] [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: 05/30/2024] [Accepted: 08/05/2024] [Indexed: 09/18/2024] Open
Abstract
Introduction Actin-binding proteins (ABPs) are essential for the regulation of morphological plasticity required for tumor cells to metastasize. The aim of this study was to perform an unbiased bioinformatic approach to identify the key ABPs significantly associated with the metastatic potential of breast cancer cells. Methods Microarray data from 181 primary breast cancer samples from our hospital were used, and all genes belonging to the Gene Ontology term actin cytoskeleton organization were obtained from QuickGO. Association with metastasis-free survival probability was tested using Cox proportional hazards regression, and pairwise co-expression was tested by Pearson correlations. Differential expression between different subgroups was analyzed using Wilcoxon tests for dichotomous traits and Kruskal-Wallis tests for categorical traits. Validation was performed using four publicly available breast cancer datasets. Results ARHGAP25 was significantly associated with a low metastatic potential, and CFL1, TMSB15A, and ACTL8 were significantly associated with a high metastatic potential. A significantly higher expression of CFL1, TMSB15A, and ACTL8 mRNA was found in the more aggressive Her2-positive and triple-negative subtypes as well as in ER-negative samples. Also, these genes were co-expressed in the same tumors. However, only mRNA levels of CFL1 were increased in pN1 compared to pN0 patients. External validation revealed that CFL1 and TMSB15A had significant associations with consistent hazard ratios in two breast cancer cohorts, and among these, CFL1 exhibited the highest hazard ratios. Conclusion CFL1 showed the strongest correlation with the metastatic potential of breast tumors. Thus, targeted inhibition of CFL1 might be a promising approach to treat malignant breast cancer cells.
Collapse
Affiliation(s)
- Christian Müller
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Volkmar Müller
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Barbara Schmalfeldt
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sabine Windhorst
- Department of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
7
|
Ying Y, Tao N, Zhang F, Wen X, Zhou M, Gao J. Thymosin β4 Regulates the Differentiation of Thymocytes by Controlling the Cytoskeletal Rearrangement and Mitochondrial Transfer of Thymus Epithelial Cells. Int J Mol Sci 2024; 25:1088. [PMID: 38256161 PMCID: PMC10816181 DOI: 10.3390/ijms25021088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/11/2024] [Accepted: 01/14/2024] [Indexed: 01/24/2024] Open
Abstract
The thymus is one of the most crucial immunological organs, undergoing visible age-related shrinkage. Thymic epithelial cells (TECs) play a vital role in maintaining the normal function of the thymus, and their degeneration is the primary cause of age-induced thymic devolution. Thymosin β4 (Tβ4) serves as a significant important G-actin sequestering peptide. The objective of this study was to explore whether Tβ4 influences thymocyte differentiation by regulating the cytoskeletal rearrangement and mitochondrial transfer of TECs. A combination of H&E staining, immunofluorescence, transmission electron microscopy, RT-qPCR, flow cytometry, cytoskeletal immunolabeling, and mitochondrial immunolabeling were employed to observe the effects of Tβ4 on TECs' skeleton rearrangement, mitochondrial transfer, and thymocyte differentiation. The study revealed that the Tβ4 primarily regulates the formation of microfilaments and the mitochondrial transfer of TECs, along with the formation and maturation of double-negative cells (CD4-CD8-) and CD4 single-positive cells (CD3+TCRβ+CD4+CD8-) thymocytes. This study suggests that Tβ4 plays a crucial role in thymocyte differentiation by influencing the cytoskeletal rearrangement and mitochondrial transfer of TECs. These effects may be associated with Tβ4's impact on the aggregation of F-actin. This finding opens up new avenues for research in the field of immune aging.
Collapse
Affiliation(s)
| | | | | | | | | | - Jianli Gao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.Y.); (N.T.); (F.Z.); (X.W.); (M.Z.)
| |
Collapse
|
8
|
Yang Z, Luo J, Zhang M, Zhan M, Bai Y, Yang Y, Wang W, Lu L. TMSB4X: A novel prognostic marker for non-small cell lung cancer. Heliyon 2023; 9:e21505. [PMID: 38027718 PMCID: PMC10663839 DOI: 10.1016/j.heliyon.2023.e21505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Non-small cell lung cancer (NSCLC), as the main type of lung cancer, has a long history of high incidence and mortality. Despite the continuous updates to the American Joint Committee on Cancer (AJCC) staging system, which adapt to evolving treatment modalities and diagnostic advancements, it is evident that patients at the same stage exhibit varying prognoses. The heterogeneity of tumors underscores the need for molecular diagnostics to assume a pivotal role in tumor staging and patient stratification. In our investigation, we meticulously analyzed the data of the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) database, incorporating clinical patients and scrutinizing pathological specimens. Through this comprehensive approach, we established a correlation between the expression of the Thymosin beta 4 X-linked (TMSB4X) gene and poorer disease-free survival (DFS) and overall survival (OS) post-surgery. Compared to the TMSB4X positive expression group, patients in the negative expression group had a better prognosis, with longer DFS (median disease-free survival (median DFS): 16.2 months vs. 11.3 months, P = 0.032) and OS (median overall survival (mOS): 29.8 months vs. 18.5 months, P = 0.033). Furthermore, our findings suggest that TMSB4X may facilitate immune evasion in non-small cell lung cancer cells by influencing the activation of infiltrating dendritic cells (DCs) in tumor infiltrating immune cells (TIICs) (R = 0.27, P = 4.8E+08). In summary, TMSB4X emerges as an unfavorable prognostic factor for NSCLC, potentially modulating the tumor immune microenvironment through its regulatory impact on dendritic cell function, thus facilitating tumor immune escape.
Collapse
Affiliation(s)
- Ze Yang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, China
- The Second Affiliated Hospital of Zunyi Medical University, Zun Yi, Gui Zhou, China
| | - Jihang Luo
- Affiliated Hospital of Zunyi Medical University, Zun Yi, Gui Zhou, China
| | - Mengmei Zhang
- Zunyi Medical and Pharmaceutical College, Zun Yi, Gui Zhou, China
| | - Meixiao Zhan
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, China
| | - Yuju Bai
- The Second Affiliated Hospital of Zunyi Medical University, Zun Yi, Gui Zhou, China
| | - Yi Yang
- The Second Affiliated Hospital of Zunyi Medical University, Zun Yi, Gui Zhou, China
| | - Wei Wang
- Department of Pulmonary and Critical Care Medicine, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, China
| | - Ligong Lu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, China
| |
Collapse
|
9
|
Myers BL, Brayer KJ, Paez-Beltran LE, Keith MS, Suzuki H, Newville J, Anderson RH, Lo Y, Mertz CM, Kollipara R, Borromeo MD, Bachoo RM, Johnson JE, Vue TY. Glioblastoma initiation, migration, and cell types are regulated by core bHLH transcription factors ASCL1 and OLIG2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.30.560206. [PMID: 37873200 PMCID: PMC10592871 DOI: 10.1101/2023.09.30.560206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Glioblastomas (GBMs) are highly aggressive, infiltrative, and heterogeneous brain tumors driven by complex driver mutations and glioma stem cells (GSCs). The neurodevelopmental transcription factors ASCL1 and OLIG2 are co-expressed in GBMs, but their role in regulating the heterogeneity and hierarchy of GBM tumor cells is unclear. Here, we show that oncogenic driver mutations lead to dysregulation of ASCL1 and OLIG2, which function redundantly to initiate brain tumor formation in a mouse model of GBM. Subsequently, the dynamic levels and reciprocal binding of ASCL1 and OLIG2 to each other and to downstream target genes then determine the cell types and degree of migration of tumor cells. Single-cell RNA sequencing (scRNA-seq) reveals that a high level of ASCL1 is key in defining GSCs by upregulating a collection of ribosomal protein, mitochondrial, neural stem cell (NSC), and cancer metastasis genes - all essential for sustaining the high proliferation, migration, and therapeutic resistance of GSCs.
Collapse
|
10
|
Kim YS, Kim D, Park J, Chung YJ. Single-cell RNA sequencing of a poorly metastatic melanoma cell line and its subclones with high lung and brain metastasis potential reveals gene expression signature of metastasis with prognostic implication. Exp Dermatol 2023; 32:1774-1784. [PMID: 37534569 DOI: 10.1111/exd.14900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 07/03/2023] [Accepted: 07/19/2023] [Indexed: 08/04/2023]
Abstract
The molecular mechanisms underlying melanoma metastasis remain poorly understood. In this study, we aimed to delineate the mechanisms underlying gene expression alterations during metastatic potential acquisition and characterize the metastatic subclones within primary cell lines. We performed single-cell RNA sequencing of a poorly metastatic melanoma cell line (WM239A) and its subclones with high metastatic potential to the lung (113/6-4L) and the brain (131/4-5B1 and 131/4-5B2). Unsupervised clustering of 8173 melanoma cells identified three distinct clusters according to cell type ('Primary', 'Lung' and 'Brain' clusters) with differential expression of MITF and AXL pathways and putative cancer and cell cycle drivers, with the lung cluster expressing intermediate but distinct gene profiles between primary and brain clusters. Principal component (PC) analysis revealed that PC2 (the second PC), which was positively associated with MITF expression and negatively with AXL pathways, primarily segregated cell types, in addition to PC1 of the cell cycle pathway. Pseudotime trajectory and RNA velocity analyses suggested the existence of cellular subsets with metastatic potential in the Primary cluster and an association between PC2 signature alteration and metastasis potential acquisition. Analysis of The Cancer Genome Atlas melanoma samples by clustering into PC2-high and -low clusters by quartiles of PC2 signature expression revealed that the PC2-high cluster was an independent significant factor for poor prognosis (p-value = 0.003) with distinct genomic and transcriptomic characteristics, compared to the PC2-low cluster. In conclusion, we identified signatures of melanoma metastasis with prognostic significance and putative pro-metastatic subclones within a primary cell line.
Collapse
Affiliation(s)
- Yoon-Seob Kim
- Department of Dermatology, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dokyeong Kim
- Department of Microbiology, IRCGP, Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Junseong Park
- Department of Microbiology, IRCGP, Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yeun-Jun Chung
- Department of Microbiology, IRCGP, Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
11
|
Wu A, Mazurkiewicz E, Donizy P, Kotowski K, Pieniazek M, Mazur AJ, Czogalla A, Trombik T. ABCA1 transporter promotes the motility of human melanoma cells by modulating their plasma membrane organization. Biol Res 2023; 56:32. [PMID: 37312227 DOI: 10.1186/s40659-023-00443-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 05/31/2023] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND Melanoma is one of the most aggressive and deadliest skin tumor. Cholesterol content in melanoma cells is elevated, and a portion of it accumulates into lipid rafts. Therefore, the plasma membrane cholesterol and its lateral organization might be directly linked with tumor development. ATP Binding Cassette A1 (ABCA1) transporter modulates physico-chemical properties of the plasma membrane by modifying cholesterol distribution. Several studies linked the activity of the transporter with a different outcome of tumor progression depending on which type. However, no direct link between human melanoma progression and ABCA1 activity has been reported yet. METHODS An immunohistochemical study on the ABCA1 level in 110 patients-derived melanoma tumors was performed to investigate the potential association of the transporter with melanoma stage of progression and prognosis. Furthermore, proliferation, migration and invasion assays, extracellular-matrix degradation assay, immunochemistry on proteins involved in migration processes and a combination of biophysical microscopy analysis of the plasma membrane organization of Hs294T human melanoma wild type, control (scrambled), ABCA1 Knockout (ABCA1 KO) and ABCA1 chemically inactivated cells were used to study the impact of ABCA1 activity on human melanoma metastasis processes. RESULTS The immunohistochemical analysis of clinical samples showed that high level of ABCA1 transporter in human melanoma is associated with a poor prognosis. Depletion or inhibition of ABCA1 impacts invasion capacities of aggressive melanoma cells. Loss of ABCA1 activity partially prevented cellular motility by affecting active focal adhesions formation via blocking clustering of phosphorylated focal adhesion kinases and active integrin β3. Moreover, ABCA1 activity regulated the lateral organization of the plasma membrane in melanoma cells. Disrupting this organization, by increasing the content of cholesterol, also blocked active focal adhesion formation. CONCLUSION Human melanoma cells reorganize their plasma membrane cholesterol content and organization via ABCA1 activity to promote motility processes and aggressiveness potential. Therefore, ABCA1 may contribute to tumor progression and poor prognosis, suggesting ABCA1 to be a potential metastatic marker in melanoma.
Collapse
Affiliation(s)
- Ambroise Wu
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383, Wrocław, Poland
| | - Ewa Mazurkiewicz
- Department of Cell Pathology, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383, Wrocław, Poland
| | - Piotr Donizy
- Department of Clinical and Experimental Pathology, Wrocław Medical University, Borowska 213, 50-556, Wrocław, Poland
| | - Krzysztof Kotowski
- Department of Clinical and Experimental Pathology, Wrocław Medical University, Borowska 213, 50-556, Wrocław, Poland
| | - Małgorzata Pieniazek
- Department of Oncology and Division of Surgical Oncology, Wrocław Medical University, Pl. Hirszfelda 12, 53-413, Wrocław, Poland
| | - Antonina J Mazur
- Department of Cell Pathology, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383, Wrocław, Poland.
| | - Aleksander Czogalla
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383, Wrocław, Poland.
| | - Tomasz Trombik
- Department of Biophysics, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383, Wrocław, Poland.
- Department of Biochemistry and Molecular Biology, Faculty of Medical Sciences, Medical University of Lublin, Chodzki 1, 20-093, Lublin, Poland.
| |
Collapse
|
12
|
Hollar D. Survey of genomic and physiological characteristics for survival in lymphoma: The NCI genomic data portal. Curr Probl Cancer 2023; 47:100955. [PMID: 36913744 DOI: 10.1016/j.currproblcancer.2023.100955] [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: 10/13/2022] [Revised: 12/31/2022] [Accepted: 02/09/2023] [Indexed: 03/07/2023]
Abstract
Lymphoma represents a myriad collection of neoplasms that impact lymphocytes. This cancer often involves disrupted cytokine, immune surveillance, and gene regulatory signaling, sometimes with expression of Epstein Barr Virus (EBV). We explored mutation patterns for People experiencing Lymphoma (PeL) in the National Cancer Institute (NCI) Genomic Data Commons (GDC), which contains detailed, deidentified genomic data on 86,046 people who have/had cancer with 2,730,388 distinctive mutations in 21,773 genes. The database included information on 536 (PeL), with the primary focal sample being the n = 30 who had complete mutational genomic data. We used correlations, independent samples t-tests, and linear regression to compare PeL demographics and vital status on mutation numbers, BMI, and mutation deleterious score across functional categories of 23 genes. PeL demonstrated varied patterns of mutated genes, consistent with most other cancer types. The primary PeL gene mutations clustered around five functional protein groups: transcriptional regulatory proteins, TNF/NFKB and cell signaling regulators, cytokine signaling proteins, cell cycle regulators, and immunoglobulins. Diagnosis Age, Birth Year, and BMI negatively (P < 0.05) correlated with Days to Death, and cell cycle mutations negatively correlated (P = 0.004) with survival days (R2 = 0.389). There were commonalities in some PeL for mutations across other cancer types based upon large sequence length, but also for 6 small cell lung cancer genes. Immunoglobulin mutations were prevalent but not for all cases. Research indicates a need for greater personalized genomics and multi-level systems analysis to evaluate facilitators and barriers for lymphoma survival.
Collapse
Affiliation(s)
- David Hollar
- Department of Community Medicine, Mercer University School of Medicine, Macon, GA.
| |
Collapse
|
13
|
Ouban A, Ameer OZ, Quek KJ, Arafah MA, Raddaoui L. Detection of Increased Expression of Claudin-1 in Triple-Negative Breast Cancer: Analysis and Clinical-Pathological Correlation. Cureus 2023; 15:e36648. [PMID: 37102018 PMCID: PMC10123009 DOI: 10.7759/cureus.36648] [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] [Accepted: 03/24/2023] [Indexed: 04/28/2023] Open
Abstract
Background Triple-negative breast cancer (TNBC) is a highly aggressive disease that lacks therapeutic targets and prognostic biomarkers. Claudin-1 is a well-described tight junction protein with prognostic value in many human cancers. Aims The need for the discovery of biomarkers of TNBC disease was a major reason for this study. Claudin-1 is a tight junction protein that has shown promising results in the prognosis and management of cancer in general. In the breast, claudin-1 expression and significance have shown variable results, especially in TNBC patients. Our study assessed expression of claudin-1 in a group of TNBC patients, and correlated this expression with clinical-pathological parameters, and with the expression of β-catenin. Materials and methods Tissues from a group of 52 TNBC patients were retrieved from the archives of the community hospital. All related information including demographical, pathologic and clinical data were retrieved. Immunohistochemistry assays of a rabbit polyclonal antibody anti-human claudin-1 were applied using the avidin-biotin peroxidase methodology. Results A statistically significant majority of TNBC cases positively expressed claudin-1 (81%, χ2=13.705; p<0.001). Most TNBC cases had grade 2 β-catenin expression (77.5%; p<0.001), and positive expression for claudin-1 correlated with that of β-catenin (χ2= 23.757; p<0.001). Claudin-1 and β-catenin expressions within tumour cells shared several features including absent or weakness of membranous expression, and redistribution of both proteins to the cytoplasm of tumour cells, and in some cases to the nuclei of these cells. Claudin-1 expression also correlates with adverse survival outcomes, where only four of 20 claudin-1-positive patients who received neo-adjuvant chemotherapy (NAC) achieved pathological complete response (pCR). Conclusions The above presents a complex role of claudin-1 in TNBC patients. In this study, claudin-1 expression was associated with poor prognostic features including invasion, metastases and adverse clinical outcomes. Claudin-1 expression in TNBC correlated with the expression of β-catenin, an important oncogene and a major contributor to the epithelial mesenchymal transition (EMT) phenomenon. Overall, the above results may serve as an impetus for further mechanistic studies to assess the exact role of claudin-1 in TNBC and its possible use in the management of this subset of breast cancer.
Collapse
Affiliation(s)
- Abderrahman Ouban
- Pathology and Molecular Medicine, Alfaisal University College of Medicine, Riyadh, SAU
| | - Omar Z Ameer
- Pharmaceutical Sciences, Alfaisal University College of Pharmacy, Riyadh, SAU
| | - Ko Jin Quek
- Family Medicine, Faculty of Biomedical Sciences, Macquarie University, Sydney, AUS
| | - Maria A Arafah
- Pathology and Laboratory Medicine, King Saud University, Riyadh, SAU
| | - Layla Raddaoui
- Oncology, Alfaisal University College of Medicine, Riyadh, SAU
| |
Collapse
|
14
|
Zhang X, Han J, Fan D, Wang J, Lin X, Zhang H, Zhang C, Bai J, Huang H, Gu Y. Lysine-40 succinylation of TAGLN2 induces glioma angiogenesis and tumor growth through regulating TMSB4X. Cancer Gene Ther 2023; 30:172-181. [PMID: 36131066 DOI: 10.1038/s41417-022-00534-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 08/12/2022] [Accepted: 09/06/2022] [Indexed: 01/19/2023]
Abstract
Protein lysine succinylation (Ksucc) represents an important regulatory mechanism of tumor development. In this work, the difference of protein Ksucc between HCMEC/D3 co-cultured with U87 (glioma endothelia cells, GEC) and without U87 (normal endothelia cells, NEC) was investigated using TMT labeling and affinity enrichment followed by high-resolution LC-MS/MS analysis. Interestingly, TAGLN2 was highly succinylated at K40 in GEC (15.36 folds vs. NEC). Compared to the Vector group, TAGLN2WT and a succinylation-mimetic TAGLN2K40E greatly promoted the angiogenesis of glioma in vitro and in vivo. Furthermore, the adhesion and metastasis of U87 co-cultured with GEC in the TAGLN2WT or TAGLN2K40E group were also significantly promoted. This was consistent with the increased expression of VE-cadherin and actin cytoskeleton remodeling induced by TAGLN2 K40succ in GEC. In addition, high K40succ of TAGLN2 was associated with poor prognosis in patients with glioma. Overexpression of TAGLN2K40E also markedly promoted the proliferation and migration of glioma cells, further analysis of in vivo xenograft tumors showed that there was a significant decrease in tumor size and angiogenesis in the TAGLN2K40R group. Notably, the co-localization of TMSB4X and TAGLN2 mainly in the nucleus and cytoplasm of glioma cells was detected by immunofluorescence staining. We identified TMSB4X as a potential target of TAGLN2, which was proved to interact with TAGLN2WT rather than TAGLN2K40A. And the inhibition of TMSB4X could markedly attenuate the proliferation and migration of glioma cells induced by TAGLN2 K40succ. The results revealed K40succ of TAGLN2 could be a novelty diagnosis and therapeutic target for gliomas.
Collapse
Affiliation(s)
- Xiaoyi Zhang
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, P. R. China
| | - Jin Han
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, P. R. China
| | - Di Fan
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, P. R. China
| | - Jiahong Wang
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, P. R. China
| | - Xiangdan Lin
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, P. R. China
| | - Hong Zhang
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, P. R. China
| | - Cai Zhang
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, P. R. China
| | - Jialing Bai
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, P. R. China
| | - Hailan Huang
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, P. R. China
| | - Yanting Gu
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, P. R. China.
| |
Collapse
|
15
|
Xiong Y, Qi Y, Pan Z, Wang S, Li B, Feng B, Xue H, Zhao R, Li G. Pancancer landscape analysis of the thymosin family identified TMSB10 as a potential prognostic biomarker and immunotherapy target in glioma. Cancer Cell Int 2022; 22:294. [PMID: 36163046 PMCID: PMC9513945 DOI: 10.1186/s12935-022-02698-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022] Open
Abstract
Background Thymosin family genes (TMSs), biologically important peptides with diverse intracellular and extracellular functions, have been shown to promote the progression of multiple cancers. However, multiomics characterization of TMSs and their role in human cancer prognosis has not been systematically performed. Methods We performed a comprehensive analysis of TMSs and thymosin β10 (TMSB10) using multiomics data from more than 10,000 tumor samples of 33 cancer types from The Cancer Genome Atlas (TCGA). We used single-sample gene set enrichment analysis (ssGSEA) and the gene set variation analysis (GSVA) algorithm to investigate the differences in tumor microenvironment (TME) cell infiltration and functional annotation for individual tumor samples, respectively. The role of TMSB10 in the malignant progression of glioma, the promotion of macrophage infiltration,and immunosuppressive polarization, and the combination drug efficacy were assessed via biological function assays. Results We comprehensively assessed genomic mutations, expression dysregulation, prognosis and immunotherapeutic response across 33 human cancer samples and showed that TMSB10 is specifically overexpressed in almost all types of cancer tissues. Further pan-cancer analysis showed that TMSB10 is closely related to the biological function, immune regulation and prognosis of glioma. Similar results were also found in several public glioma cohorts and our Qilu local cohort. Further integration with other biological experiments revealed the key roles of TMSB10 in the malignant progression of glioma, the promotion of macrophage infiltration and immunosuppressive polarization. We also identified multiple drugs targeting cells with high TMSB10 expression and validated that knockdown of TMSB10 improved the efficacy of selumetinib (a MEK1/2 inhibitor approved by the FDA for the treatment of neurofibromatosis-associated tumors) and anti-PD1 treatment in glioma. Conclusion These results indicate that TMSB10 holds promise as a novel prognostic marker and therapeutic target, providing a theoretical basis for the development of more effective and targeted clinical treatment strategies for glioma patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02698-5.
Collapse
Affiliation(s)
- Ye Xiong
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012, Shandong, China.,Department of Neurosurgery, The Frist Affiliated Hospital of Wenzhou Medical University, Wenzhou City, 325000, Zhejiang Province, China
| | - Yanhua Qi
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012, Shandong, China
| | - Ziwen Pan
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012, Shandong, China
| | - Shaobo Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012, Shandong, China
| | - Boyan Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012, Shandong, China
| | - Bowen Feng
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012, Shandong, China
| | - Hao Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China. .,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012, Shandong, China.
| | - Rongrong Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China. .,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012, Shandong, China.
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China. .,Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250012, Shandong, China.
| |
Collapse
|
16
|
Sun C, Zou H, Yang Z, Yang M, Chen X, Huang Y, Fan W, Yuan R. Proteomics and phosphoproteomics analysis of vitreous in idiopathic epiretinal membrane patients. Proteomics Clin Appl 2022; 16:e2100128. [PMID: 35510950 DOI: 10.1002/prca.202100128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 03/18/2022] [Accepted: 05/02/2022] [Indexed: 12/30/2022]
Abstract
PURPOSE The purpose of the present study was to characterize the idiopathic epiretinal membrane (iERM) through proteomics and phosphoproteomics analysis to facilitate the diagnosis and treatment of iERM. EXPERIMENTAL DESIGN The vitreous of 25 patients with an iERM and 15 patients with an idiopathic macular hole were analyzed by proteomic and phosphoproteomic analysis based on tandem mass tag. PRM was used to verify the differential proteins. RESULTS Proteomic analysis identified a total of 878 proteins, including 50 differential proteins. Tenascin-C, galectin-3-binding protein, glucose-6-phosphate isomerase, neuroserpin, collagen alpha-1(XI) chain, and collagen alpha-1(II) chain were verified to be upregulated in iERM by PRM. Phosphoproteomic analysis identified a total of 401 phosphorylation sites on 213 proteins, including 27 differential phosphorylation sites on 24 proteins. Mitogen-activated protein kinase-activated protein kinase (MAPKAPK)3 and MAPKAPK5 were predicted as the major kinases in the vitreous of iERM. Twenty-six of the differential proteins and phosphorylated proteins may be closely related to fibrosis in iERM. CONCLUSION AND CLINICAL RELEVANCE Our results indicated the potential biomarkers or therapeutic targets for iERM, provided key kinases that may be involved in iERM. Fibrosis plays an essential role in iERM, and further exploration of related differential proteins has important clinical significance.
Collapse
Affiliation(s)
- Chao Sun
- Department of Ophthalmology, the Second Affiliated Hospital of Army Medical University, Chongqing, PR China
| | - Huan Zou
- Department of Ophthalmology, the Second Affiliated Hospital of Army Medical University, Chongqing, PR China
| | - Zhouquan Yang
- Department of Ophthalmology, the Second Affiliated Hospital of Army Medical University, Chongqing, PR China
| | - Mei Yang
- Department of Ophthalmology, the Second Affiliated Hospital of Army Medical University, Chongqing, PR China
| | - Xiaofan Chen
- Department of Ophthalmology, the Second Affiliated Hospital of Army Medical University, Chongqing, PR China
| | - Yanming Huang
- Department of Ophthalmology, the Second Affiliated Hospital of Army Medical University, Chongqing, PR China
| | - Wei Fan
- Department of Ophthalmology, the Second Affiliated Hospital of Army Medical University, Chongqing, PR China
| | - Rongdi Yuan
- Department of Ophthalmology, the Second Affiliated Hospital of Army Medical University, Chongqing, PR China
| |
Collapse
|
17
|
Zou S, Zhang Y, Zhang L, Wang D, Xu S. Construction and validation of a prognostic risk model of angiogenesis factors in skin cutaneous melanoma. Aging (Albany NY) 2022; 14:1529-1548. [PMID: 35157610 PMCID: PMC8876895 DOI: 10.18632/aging.203895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 02/08/2022] [Indexed: 12/24/2022]
Abstract
Melanoma can secrete tumor angiogenesis factors, which is the essential factor for tumor growth and metastasis. However, there are few reports on the relationship between angiogenesis factors and prognosis risk in melanoma. This study aimed to develop a prognostic risk model of angiogenesis for melanoma. Forty-nine differentially expressed angiogenesis were identified from the TCGA database, which were mainly involved in PI3K/Akt pathway, focal adhesion, and MAPK signaling pathway. We then establish an eleven-gene signature. The model indicated a strong prognostic capability in both the discovery cohort and the validation cohort. Patients of smaller height (<170 cm) and lower weight (<80 kg) and those with advanced-stage and ulcerated melanoma had higher risk scores. The risk score was positively correlated with mutation load, homologous recombination defect, neoantigen load and chromosome instability. In addition, the high-risk group had a higher degree of immune cell infiltration, better response to immunotherapy and lower immune score. Therefore, these results indicate that the risk model is an effective method to predict the prognosis of melanoma.
Collapse
Affiliation(s)
- Songyun Zou
- Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong, China
| | - Yonggang Zhang
- Department of Clinical Laboratory, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong, China
| | - Limei Zhang
- Oncology Department, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong, China
| | - Dengchuan Wang
- Office of Medical Ethics, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong, China
| | - Shi Xu
- Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
18
|
Górska A, Mazur AJ. Integrin-linked kinase (ILK): the known vs. the unknown and perspectives. Cell Mol Life Sci 2022; 79:100. [PMID: 35089438 PMCID: PMC8799556 DOI: 10.1007/s00018-021-04104-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/29/2021] [Accepted: 12/17/2021] [Indexed: 02/08/2023]
Abstract
Integrin-linked kinase (ILK) is a multifunctional molecular actor in cell-matrix interactions, cell adhesion, and anchorage-dependent cell growth. It combines functions of a signal transductor and a scaffold protein through its interaction with integrins, then facilitating further protein recruitment within the ILK-PINCH-Parvin complex. ILK is involved in crucial cellular processes including proliferation, survival, differentiation, migration, invasion, and angiogenesis, which reflects on systemic changes in the kidney, heart, muscle, skin, and vascular system, also during the embryonal development. Dysfunction of ILK underlies the pathogenesis of various diseases, including the pro-oncogenic activity in tumorigenesis. ILK localizes mostly to the cell membrane and remains an important component of focal adhesion. We do know much about ILK but a lot still remains either uncovered or unclear. Although it was initially classified as a serine/threonine-protein kinase, its catalytical activity is now questioned due to structural and functional issues, leaving the exact molecular mechanism of signal transduction by ILK unsolved. While it is known that the three isoforms of ILK vary in length, the presence of crucial domains, and modification sites, most of the research tends to focus on the main isoform of this protein while the issue of functional differences of ILK2 and ILK3 still awaits clarification. The activity of ILK is regulated on the transcriptional, protein, and post-transcriptional levels. The crucial role of phosphorylation and ubiquitylation has been investigated, but the functions of the vast majority of modifications are still unknown. In the light of all those open issues, here we present an extensive literature survey covering a wide spectrum of latest findings as well as a past-to-present view on controversies regarding ILK, finishing with pointing out some open questions to be resolved by further research.
Collapse
Affiliation(s)
- Agata Górska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, ul. Joliot-Curie 14a, 50-383, Wrocław, Poland.
| | - Antonina Joanna Mazur
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, ul. Joliot-Curie 14a, 50-383, Wrocław, Poland.
| |
Collapse
|
19
|
Sun L, Wei Y, Wang J. Circular RNA PIP5K1A (circPIP5K1A) accelerates endometriosis progression by regulating the miR-153-3p/Thymosin Beta-4 X-Linked (TMSB4X) pathway. Bioengineered 2021; 12:7104-7118. [PMID: 34546850 PMCID: PMC8806837 DOI: 10.1080/21655979.2021.1978618] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
As a common gynecologic disease, endometriosis (EM) poses a threat to the reproductive health of about 10% women globally. Recent studies have revealed that circular RNAs (circRNAs) are deeply implicated in EM pathogenesis. However, the functions of circPIP5K1A in EM have not been studied yet. Our study intended to uncover the molecular mechanism of circPIP5K1A in EM. In this work, gene and protein expressions were determined by RT-qPCR or Western blotting. CCK-8, wound healing, transwell, and flow cytometry assays were conducted to analyze cell viability, migration, invasion, cell cycle, and apoptosis. Additionally, bioinformatics analysis, dual-luciferase reporter assay, as well as RIP assay were performed to investigate the combination between miR-153-3p and circPIP5K1A or TMSB4X. Herein, we found remarkable high circPIP5K1A expression in EM tissues and cells. Silencing of circPIP5K1A suppressed proliferation, restrained cell cycle, increased cell apoptosis, and decreased migration and invasion in EM cells. In addition, miR-153-3p inhibition could abrogate the impacts of circPIP5K1A knockdown on EM progression in vitro. Also, we found that circPIP5K1A regulated TMSB4X level via interaction with miR-153-3p in EM cells. Besides, circPIP5K1A promoted EM progression via TMSB4X. Moreover, TMSB4X could activate the TGF-β signaling in hEM15A cells. To sum up, our study elucidated that circPIP5K1A accelerated EM progression in vitro by activating the TGF-β signaling pathway via the miR-153-3p/TMSB4X axis, providing a potential clinical target for EM treatment.
Collapse
Affiliation(s)
- Lin Sun
- Department of Gynecology, Maanshan Maternal and Child Health Care Hospital, Ma'anshan, Anhui, P.R.China
| | - Yan Wei
- Department of Gynecology, The Affiliated Suzhou Science&Technology Town Hospital of Nanjing Medical University, Suzhou, Jiangsu, P.R.China
| | - Junli Wang
- Department of Gynecology, Maanshan Maternal and Child Health Care Hospital, Ma'anshan, Anhui, P.R.China
| |
Collapse
|
20
|
Effectiveness of bortezomib and temozolomide for eradication of recurrent human glioblastoma cells, resistant to radiation. PROGRESS IN BRAIN RESEARCH 2021; 266:195-209. [PMID: 34689859 DOI: 10.1016/bs.pbr.2021.06.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is a primary human brain tumor with the highest mortality rate. The prognosis for such patients is unfavorable, since the tumor is highly resistant to treatment, and the median survival of patients is 13 months. Chemotherapy might extend patients' life, but a tumor, that reappears after chemoradiotherapy, is resistant to temozolomide (TMZ). Using postgenome technologies in clinical practice might have a positive effect on the treatment of a recurrent GBM. METHODS T98G cells of human GBM have been used. Radiation treatment was performed with Rokus-M gamma-therapeutic system, using 60Сo as a source of radionuclide emissions. High-performance liquid chromatography-mass spectrometry was used for proteome analysis. Mass spectrometry data were processed with MaxQuant (version 1.6.1.0) and Perseus (version 1.6.1) software, Max Planck Institute of Biochemistry (Germany). Biological processes, molecular functions, cells locations and protein pathways were annotated with a help of PubMed, PANTHER, Gene Ontology and KEGG and STRING v10 databases. Pharmaceutical testing was performed in vitro with a panel of traditional chemotherapeutic agents. RESULTS GBM cells proliferation speed is inversely proportional to the irradiation dose and recedes when the dosage is increased, as expected. Synthesis of ERC1, NARG1L, PLCD3, ROCK2, SARNP, TMSB4X and YTHDF2 in GBM cells, treated with 60Gy of radiation, shows more than a fourfold increase, while the synthesis level of PSMA2, PSMA3, PSMA4, PSMB2, PSMB3, PSMB7, PSMC3, PSMD1, PSMD3 proteins increases significantly. Traditional chemotherapeutic agents are not very effective against cancer cells of the recurrent GBM. Combination of TMZ and CCNU with a proteasome inhibitor-bortezomib-significantly increases their ability to eradicate cells of a radioresistant GBM. CONCLUSIONS Bortezomib and temozolomide effectively destroy cells of a radioresistant recurrent human glioblastoma; proteome mapping of the recurrent GBM cancer cells allows to identify new targets for therapy to improve the treatment results.
Collapse
|
21
|
Chi LH, Wu ATH, Hsiao M, Li YC(J. A Transcriptomic Analysis of Head and Neck Squamous Cell Carcinomas for Prognostic Indications. J Pers Med 2021; 11:782. [PMID: 34442426 PMCID: PMC8399099 DOI: 10.3390/jpm11080782] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 01/27/2023] Open
Abstract
Survival analysis of the Cancer Genome Atlas (TCGA) dataset is a well-known method for discovering gene expression-based prognostic biomarkers of head and neck squamous cell carcinoma (HNSCC). A cutoff point is usually used in survival analysis for patient dichotomization when using continuous gene expression values. There is some optimization software for cutoff determination. However, the software's predetermined cutoffs are usually set at the medians or quantiles of gene expression values. There are also few clinicopathological features available in pre-processed datasets. We applied an in-house workflow, including data retrieving and pre-processing, feature selection, sliding-window cutoff selection, Kaplan-Meier survival analysis, and Cox proportional hazard modeling for biomarker discovery. In our approach for the TCGA HNSCC cohort, we scanned human protein-coding genes to find optimal cutoff values. After adjustments with confounders, clinical tumor stage and surgical margin involvement were found to be independent risk factors for prognosis. According to the results tables that show hazard ratios with Bonferroni-adjusted p values under the optimal cutoff, three biomarker candidates, CAMK2N1, CALML5, and FCGBP, are significantly associated with overall survival. We validated this discovery by using the another independent HNSCC dataset (GSE65858). Thus, we suggest that transcriptomic analysis could help with biomarker discovery. Moreover, the robustness of the biomarkers we identified should be ensured through several additional tests with independent datasets.
Collapse
Affiliation(s)
- Li-Hsing Chi
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan; (L.-H.C.); (A.T.H.W.)
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Wan Fang Hospital, Taipei Medical University, Taipei 11600, Taiwan
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
| | - Alexander T. H. Wu
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan; (L.-H.C.); (A.T.H.W.)
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei 115024, Taiwan
- Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Yu-Chuan (Jack) Li
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan; (L.-H.C.); (A.T.H.W.)
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, No.172-1, Sec. 2, Keelung Rd., Taipei 106339, Taiwan
| |
Collapse
|
22
|
Bannerman D, Pascual-Gil S, Floryan M, Radisic M. Bioengineering strategies to control epithelial-to-mesenchymal transition for studies of cardiac development and disease. APL Bioeng 2021; 5:021504. [PMID: 33948525 PMCID: PMC8068500 DOI: 10.1063/5.0033710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 03/15/2021] [Indexed: 12/24/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a process that occurs in a wide range of tissues and environments, in response to numerous factors and conditions, and plays a critical role in development, disease, and regeneration. The process involves epithelia transitioning into a mobile state and becoming mesenchymal cells. The investigation of EMT processes has been important for understanding developmental biology and disease progression, enabling the advancement of treatment approaches for a variety of disorders such as cancer and myocardial infarction. More recently, tissue engineering efforts have also recognized the importance of controlling the EMT process. In this review, we provide an overview of the EMT process and the signaling pathways and factors that control it, followed by a discussion of bioengineering strategies to control EMT. Important biological, biomaterial, biochemical, and physical factors and properties that have been utilized to control EMT are described, as well as the studies that have investigated the modulation of EMT in tissue engineering and regenerative approaches in vivo, with a specific focus on the heart. Novel tools that can be used to characterize and assess EMT are discussed and finally, we close with a perspective on new bioengineering methods that have the potential to transform our ability to control EMT, ultimately leading to new therapies.
Collapse
|
23
|
Zhu Y, Lesch A, Li X, Lin TE, Gasilova N, Jović M, Pick HM, Ho PC, Girault HH. Rapid Noninvasive Skin Monitoring by Surface Mass Recording and Data Learning. JACS AU 2021; 1:598-611. [PMID: 34056635 PMCID: PMC8154208 DOI: 10.1021/jacsau.0c00074] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Indexed: 05/08/2023]
Abstract
Skin problems are often overlooked due to a lack of robust and patient-friendly monitoring tools. Herein, we report a rapid, noninvasive, and high-throughput analytical chemical methodology, aiming at real-time monitoring of skin conditions and early detection of skin disorders. Within this methodology, adhesive sampling and laser desorption ionization mass spectrometry are coordinated to record skin surface molecular mass in minutes. Automated result interpretation is achieved by data learning, using similarity scoring and machine learning algorithms. Feasibility of the methodology has been demonstrated after testing a total of 117 healthy, benign-disordered, or malignant-disordered skins. Remarkably, skin malignancy, using melanoma as a proof of concept, was detected with 100% accuracy already at early stages when the lesions were submillimeter-sized, far beyond the detection limit of most existing noninvasive diagnosis tools. Moreover, the malignancy development over time has also been monitored successfully, showing the potential to predict skin disorder progression. Capable of detecting skin alterations at the molecular level in a nonsurgical and time-saving manner, this analytical chemistry platform is promising to build personalized skin care.
Collapse
Affiliation(s)
- Yingdi Zhu
- Institute of Chemical Sciences and Engineering, School of Basic Sciences, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Andreas Lesch
- Department of Industrial Chemistry "Toso Montanari", Universita degli Studi di Bologna, 40136 Bologna, Italy
| | - Xiaoyun Li
- Department of Fundamental Oncology, Université de Lausanne, 1066 Epalinges, Switzerland
- Ludwig Institute for Cancer Research, Université de Lausanne, 1066 Epalinges, Switzerland
| | - Tzu-En Lin
- Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, 30010 Hsinchu, Taiwan
| | - Natalia Gasilova
- Institute of Chemical Sciences and Engineering, School of Basic Sciences, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Milica Jović
- Institute of Chemical Sciences and Engineering, School of Basic Sciences, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Horst Matthias Pick
- Environmental Engineering Institute, School of Architecture, Civil and Environmental Engineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Ping-Chih Ho
- Department of Fundamental Oncology, Université de Lausanne, 1066 Epalinges, Switzerland
- Ludwig Institute for Cancer Research, Université de Lausanne, 1066 Epalinges, Switzerland
| | - Hubert H Girault
- Institute of Chemical Sciences and Engineering, School of Basic Sciences, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| |
Collapse
|
24
|
Makowiecka A, Mazurkiewicz E, Mrówczyńska E, Malek N, Battistella A, Lazzarino M, Nowak D, Mazur AJ. Changes in Biomechanical Properties of A375 Cells Due to the Silencing of TMSB4X Expression Are Not Directly Correlated with Alterations in Their Stemness Features. Cells 2021; 10:cells10040769. [PMID: 33807338 PMCID: PMC8067020 DOI: 10.3390/cells10040769] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/24/2021] [Accepted: 03/28/2021] [Indexed: 12/27/2022] Open
Abstract
Thymosin β4 (Tβ4) is a small, 44-amino acid polypeptide. It has been implicated in multiple processes, including cell movement, angiogenesis, and stemness. Previously, we reported that melanoma cell lines differ in Tβ4 levels. Studies on stable clones with silenced TMSB4X expression showed that Tβ4 impacted adhesion and epithelial-mesenchymal transition progression. Here, we show that the cells with silenced TMSB4X expression exhibited altered actin cytoskeleton’s organization and subcellular relocalization of two intermediate filament proteins: Nestin and Vimentin. The rearrangement of the cell cytoskeleton resulted in changes in the cells’ topology, height, and stiffness defined by Young’s modulus. Simultaneously, only for some A375 clones with a lowered Tβ4 level, we observed a decreased ability to initiate colony formation in soft agar, tumor formation in vivo, and alterations in Nanog’s expression level transcription factor regulating stemness. Thus, we show for the first time that in A375 cells, biomechanical properties are not directly coupled to stemness features, and this cell line is phenotypically heterogeneous.
Collapse
Affiliation(s)
- Aleksandra Makowiecka
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, 50-383 Wrocław, Poland; (E.M.); (E.M.); (N.M.); (D.N.)
- Istituto Officina dei Materiali-National Research Council, I-34149 Trieste, Italy; (A.B.); (M.L.)
- Correspondence: or (A.M.); (A.J.M.); Tel.: +48-71-375-6206 (A.J.M.)
| | - Ewa Mazurkiewicz
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, 50-383 Wrocław, Poland; (E.M.); (E.M.); (N.M.); (D.N.)
| | - Ewa Mrówczyńska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, 50-383 Wrocław, Poland; (E.M.); (E.M.); (N.M.); (D.N.)
| | - Natalia Malek
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, 50-383 Wrocław, Poland; (E.M.); (E.M.); (N.M.); (D.N.)
| | - Alice Battistella
- Istituto Officina dei Materiali-National Research Council, I-34149 Trieste, Italy; (A.B.); (M.L.)
| | - Marco Lazzarino
- Istituto Officina dei Materiali-National Research Council, I-34149 Trieste, Italy; (A.B.); (M.L.)
| | - Dorota Nowak
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, 50-383 Wrocław, Poland; (E.M.); (E.M.); (N.M.); (D.N.)
| | - Antonina Joanna Mazur
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, 50-383 Wrocław, Poland; (E.M.); (E.M.); (N.M.); (D.N.)
- Correspondence: or (A.M.); (A.J.M.); Tel.: +48-71-375-6206 (A.J.M.)
| |
Collapse
|
25
|
Malek N, Michrowska A, Mazurkiewicz E, Mrówczyńska E, Mackiewicz P, Mazur AJ. The origin of the expressed retrotransposed gene ACTBL2 and its influence on human melanoma cells' motility and focal adhesion formation. Sci Rep 2021; 11:3329. [PMID: 33558623 PMCID: PMC7870945 DOI: 10.1038/s41598-021-82074-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/08/2021] [Indexed: 01/30/2023] Open
Abstract
We have recently found that β-actin-like protein 2 (actbl2) forms complexes with gelsolin in human melanoma cells and can polymerize. Phylogenetic and bioinformatic analyses showed that actbl2 has a common origin with two non-muscle actins, which share a separate history from the muscle actins. The actin groups' divergence started at the beginning of vertebrate evolution, and actbl2 actins are characterized by the largest number of non-conserved amino acid substitutions of all actins. We also discovered that ACTBL2 is expressed at a very low level in several melanoma cell lines, but a small subset of cells exhibited a high ACTBL2 expression. We found that clones with knocked-out ACTBL2 (CR-ACTBL2) or overexpressing actbl2 (OE-ACTBL2) differ from control cells in the invasion, focal adhesion formation, and actin polymerization ratio, as well as in the formation of lamellipodia and stress fibers. Thus, we postulate that actbl2 is the seventh actin isoform and is essential for cell motility.
Collapse
Affiliation(s)
- Natalia Malek
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, ul. Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Aleksandra Michrowska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, ul. Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Ewa Mazurkiewicz
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, ul. Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Ewa Mrówczyńska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, ul. Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Paweł Mackiewicz
- Department of Bioinformatics and Genomics, Faculty of Biotechnology, University of Wroclaw, ul. Joliot-Curie 14a, Wroclaw, 50-383, Poland
| | - Antonina J Mazur
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, ul. Joliot-Curie 14a, 50-383, Wroclaw, Poland.
| |
Collapse
|
26
|
Abstract
Simple Summary Cell migration is an essential process from embryogenesis to cell death. This is tightly regulated by numerous proteins that help in proper functioning of the cell. In diseases like cancer, this process is deregulated and helps in the dissemination of tumor cells from the primary site to secondary sites initiating the process of metastasis. For metastasis to be efficient, cytoskeletal components like actin, myosin, and intermediate filaments and their associated proteins should co-ordinate in an orderly fashion leading to the formation of many cellular protrusions-like lamellipodia and filopodia and invadopodia. Knowledge of this process is the key to control metastasis of cancer cells that leads to death in 90% of the patients. The focus of this review is giving an overall understanding of these process, concentrating on the changes in protein association and regulation and how the tumor cells use it to their advantage. Since the expression of cytoskeletal proteins can be directly related to the degree of malignancy, knowledge about these proteins will provide powerful tools to improve both cancer prognosis and treatment. Abstract Successful metastasis depends on cell invasion, migration, host immune escape, extravasation, and angiogenesis. The process of cell invasion and migration relies on the dynamic changes taking place in the cytoskeletal components; actin, tubulin and intermediate filaments. This is possible due to the plasticity of the cytoskeleton and coordinated action of all the three, is crucial for the process of metastasis from the primary site. Changes in cellular architecture by internal clues will affect the cell functions leading to the formation of different protrusions like lamellipodia, filopodia, and invadopodia that help in cell migration eventually leading to metastasis, which is life threatening than the formation of neoplasms. Understanding the signaling mechanisms involved, will give a better insight of the changes during metastasis, which will eventually help targeting proteins for treatment resulting in reduced mortality and longer survival.
Collapse
|
27
|
Malek N, Mrówczyńska E, Michrowska A, Mazurkiewicz E, Pavlyk I, Mazur AJ. Knockout of ACTB and ACTG1 with CRISPR/Cas9(D10A) Technique Shows that Non-Muscle β and γ Actin Are Not Equal in Relation to Human Melanoma Cells' Motility and Focal Adhesion Formation. Int J Mol Sci 2020; 21:ijms21082746. [PMID: 32326615 PMCID: PMC7216121 DOI: 10.3390/ijms21082746] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/10/2020] [Accepted: 04/10/2020] [Indexed: 12/12/2022] Open
Abstract
Non-muscle actins have been studied for many decades; however, the reason for the existence of both isoforms is still unclear. Here we show, for the first time, a successful inactivation of the ACTB (CRISPR clones with inactivated ACTB, CR-ACTB) and ACTG1 (CRISPR clones with inactivated ACTG1, CR-ACTG1) genes in human melanoma cells (A375) via the RNA-guided D10A mutated Cas9 nuclease gene editing [CRISPR/Cas9(D10A)] technique. This approach allowed us to evaluate how melanoma cell motility was impacted by the lack of either β actin coded by ACTB or γ actin coded by ACTG1. First, we observed different distributions of β and γ actin in the cells, and the absence of one actin isoform was compensated for via increased expression of the other isoform. Moreover, we noted that γ actin knockout had more severe consequences on cell migration and invasion than β actin knockout. Next, we observed that the formation rate of bundled stress fibers in CR-ACTG1 cells was increased, but lamellipodial activity in these cells was impaired, compared to controls. Finally, we discovered that the formation rate of focal adhesions (FAs) and, subsequently, FA-dependent signaling were altered in both the CR-ACTB and CR-ACTG1 clones; however, a more detrimental effect was observed for γ actin-deficient cells. Our research shows that both non-muscle actins play distinctive roles in melanoma cells’ FA formation and motility.
Collapse
|