1
|
Libring S, Reinhart-King CA. Premetastatic niche mechanics and organotropism in breast cancer. NPJ BIOLOGICAL PHYSICS AND MECHANICS 2025; 2:11. [PMID: 40191104 PMCID: PMC11968405 DOI: 10.1038/s44341-025-00015-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 02/14/2025] [Indexed: 04/09/2025]
Abstract
Numerous physical and mechanical changes occur in the premetastatic niche. Here, we review the mechanics of the premetastatic niche and how the altered extracellular matrix and cancer cell mechanics may play a role in organotropism in breast cancer. Future research into premetastatic niche development and organotropic cell behavior should address physical alterations and biomechanical effects to the same rigor that biochemical alterations are studied.
Collapse
Affiliation(s)
- Sarah Libring
- Department of Bioengineering, Rice University, Houston, TX USA
| | | |
Collapse
|
2
|
Zhang H, Wang X, Dong M, Wang J, Ren W. Unveiling novel regulatory mechanisms of miR-5195-3p in pelvic organ prolapse pathogenesis†. Biol Reprod 2025; 112:86-101. [PMID: 39530351 DOI: 10.1093/biolre/ioae162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 10/09/2024] [Accepted: 11/11/2024] [Indexed: 11/16/2024] Open
Abstract
Pelvic organ prolapse is a condition that significantly affects women's quality of life. The pathological mechanism of pelvic organ prolapse is not yet fully understood, and its pathogenesis is often caused by multiple factors, including the metabolic imbalance of the extracellular matrix. This study aims to investigate the role of miR-5195-3p, a microRNA, in the pathology of pelvic organ prolapse and its regulatory mechanism. Using various molecular biology techniques such as real-time reverse transcription Polymerase Chain Reaction (PCR), fluorescence in situ hybridization, immunohistochemistry, and Western blot, miR-5195-3p expression was examined in vaginal wall tissues obtained from pelvic organ prolapse patients. Results revealed an up-regulation of miR-5195-3p expression in these tissues, showing a negative correlation with the expression of extracellular matrix-related proteins. Further analysis using bioinformatics tools identified Lipoxygenase (LOX) as a potential target in pelvic organ prolapse. Dual luciferase reporter gene experiments confirmed LOX as a direct target of miR-5195-3p. Interestingly, regulating the expression of LOX also influenced the transforming growth factor β1 signaling pathway and had an impact on extracellular matrix metabolism. This finding suggests that miR-5195-3p controls extracellular matrix metabolism by targeting LOX and modulating the TGF-β1 signaling pathway. In conclusion, this study unveils the involvement of miR-5195-3p in the pathological mechanism of pelvic organ prolapse by regulating extracellular matrix metabolism through the LOX/TGF-β1 axis. These findings reveal new mechanisms in the pathogenesis of pelvic organ prolapse, providing a theoretical foundation and therapeutic targets for further research on pelvic organ prolapse treatment.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xinlu Wang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Meng Dong
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Jie Wang
- Department of Health Management, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang 110004, China
| | - Weidong Ren
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
3
|
Li H, Chen J, Liu Z, Pan L, Lan X, Jiang L, Huang F. Construction of a novel copper-induced-cell-death-related gene signature for prognosis in colon cancer, with focus on KIF7. BMC Cancer 2024; 24:1532. [PMID: 39695482 DOI: 10.1186/s12885-024-13315-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Colon cancer (CC) is a leading cause of cancer-related mortality worldwide. Accurate prognostic markers are essential for patient risk stratification and personalized treatment. Copper-induced cell-death-related genes (CRG) have emerged as potential players in cancer prognosis, yet their role in CC remains unclear. METHODS This study aimed to comprehensively evaluate the expression of CRG and their roles in CC using gene expression and clinical data from TCGA and GEO databases. Univariate Cox regression and least absolute shrinkage and selection operator (LASSO) analyses identified prognostic genes, leading to the construction of a CRG prognostic signature. The signature's predictive accuracy was validated using Kaplan-Meier survival curves, Receiver Operating Characteristic (ROC) curves, and a nomogram model. Additionally, we conducted experiments including immunofluorescence staining and cellular assays to validate the key genes' biological functions. RESULTS A 12-gene CRG signature was significantly associated with overall survival in CC patients. The high-risk group, as classified by median risk score, exhibited significantly shorter survival times compared to the low-risk group. The signature's predictive accuracy was further confirmed with Area Under the Curve (AUC) scores exceeding 0.75 in TCGA and GSE17536 cohorts. Notably, the risk score was significantly correlated with immune checkpoints, chemotherapy sensitivity, and tumor microenvironment. Furthermore, the risk score showed a strong association with immunotherapy response in patients from GSE78220 and GSE39688 cohorts. Bioinformatics analysis of KIF7, a key gene within the signature, revealed its upregulation in CC and significant associations with tumor mutation burden, microsatellite instability, and immune cell infiltration across various cancers. Experiments confirmed that KIF7 was upregulated in CC and its knockdown reduced cell proliferation, migration, and invasion. CONCLUSION The CRG prognostic signature can effectively predict overall survival, immune microenvironment and chemotherapy response in CC. KIF7, as a potential prognostic marker, has significant potential for the prediction and treatment of CC.
Collapse
Affiliation(s)
- Hua Li
- Department of General Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, No. 18 Zhongshan 2nd Road, Baise, Guangxi, 533000, China
- Key Laboratory of Tumor Molecular Pathology of Baise, Baise, Guangxi, China
| | - Jingying Chen
- Department of General Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, No. 18 Zhongshan 2nd Road, Baise, Guangxi, 533000, China
- Graduate School, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Zhengxian Liu
- Department of General Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, No. 18 Zhongshan 2nd Road, Baise, Guangxi, 533000, China
- Graduate School, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Lujuan Pan
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Xiaoling Lan
- Key Laboratory of Tumor Molecular Pathology of Baise, Baise, Guangxi, China
| | - Lihe Jiang
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, No. 98 Chengxiang Road, Baise, Guangxi, 533000, China.
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, Fuzhou, Fujian, China.
| | - Fuda Huang
- Department of General Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, No. 18 Zhongshan 2nd Road, Baise, Guangxi, 533000, China.
- Key Laboratory of Tumor Molecular Pathology of Baise, Baise, Guangxi, China.
| |
Collapse
|
4
|
Zhu S, Jin G, He X, Li Y, Xu F, Guo H. Mechano-assisted strategies to improve cancer chemotherapy. Life Sci 2024; 359:123178. [PMID: 39471901 DOI: 10.1016/j.lfs.2024.123178] [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: 05/23/2024] [Revised: 07/25/2024] [Accepted: 10/22/2024] [Indexed: 11/01/2024]
Abstract
Chemotherapy remains a cornerstone in cancer treatment; however, its effectiveness is frequently undermined by the development of drug resistance. Recent studies underscores the pivotal role of the tumor mechanical microenvironment (TMME) and the emerging field of mechanical nanomedicine in tackling chemo-resistance. This review offers an in-depth analysis of mechano-assisted strategies aimed at mitigating chemo-resistance through the modification of the TMME and the refinement of mechanical nanomedicine delivery systems. We explore the potential of targeting abnormal tumor mechanical properties as a promising avenue for enhancing the efficacy of cancer chemotherapy, which offers novel directions for advancing future cancer therapies, especially from the mechanomedicine perspective.
Collapse
Affiliation(s)
- Shanshan Zhu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Guorui Jin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Xiaocong He
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yuan Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.
| | - Hui Guo
- Department of Medical Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, PR China.
| |
Collapse
|
5
|
Zhang X, Han X. Targeting cuproptosis for cancer therapy: Focus on the anti-tumor immune system. CANCER PATHOGENESIS AND THERAPY 2024. [DOI: 10.1016/j.cpt.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
6
|
Micalet A, Upadhyay A, Javanmardi Y, de Brito CG, Moeendarbary E, Cheema U. Patient-specific colorectal-cancer-associated fibroblasts modulate tumor microenvironment mechanics. iScience 2024; 27:110060. [PMID: 38883829 PMCID: PMC11179580 DOI: 10.1016/j.isci.2024.110060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/19/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) play a major role in reorganizing the physical tumor micro-environment and changing tissue stiffness. Herein, using an engineered three-dimensional (3D) model that mimics the tumor's native biomechanical environment, we characterized the changes in matrix stiffness caused by six patient-specific colorectal CAF populations. After 21 days of culture, atomic force microscopy (AFM) was performed to precisely measure the local changes in tissue stiffness. Each CAF population exhibited heterogeneity in remodeling capabilities, with some patient-derived cells stiffening the matrix and others softening it. Tissue stiffening was mainly attributed to active contraction of the matrix by the cells, whereas the softening was due to enzymatic activity of matrix-cleaving proteins. This measured heterogeneity was lost when the CAFs were cocultured with colorectal cancer cells, as all samples significantly soften the tissue. The interplay between cancer cells and CAFs was critical as it altered any heterogeneity exhibited by CAFs alone.
Collapse
Affiliation(s)
- Auxtine Micalet
- UCL Centre for 3D Models of Health and Disease, Department of Targeted Intervention, Division of Surgery and Interventional Science, University College London, Charles Bell House, 43-45 Foley Street, London W1W 7TS, UK
- Department of Mechanical Engineering, University College London, Gower Street, London WC1E 6BT, UK
| | - Anuja Upadhyay
- UCL Centre for 3D Models of Health and Disease, Department of Targeted Intervention, Division of Surgery and Interventional Science, University College London, Charles Bell House, 43-45 Foley Street, London W1W 7TS, UK
| | - Yousef Javanmardi
- Department of Mechanical Engineering, University College London, Gower Street, London WC1E 6BT, UK
| | | | - Emad Moeendarbary
- Department of Mechanical Engineering, University College London, Gower Street, London WC1E 6BT, UK
- 199 Biotechnologies Ltd, Gloucester Road, London W2 6LD, UK
| | - Umber Cheema
- UCL Centre for 3D Models of Health and Disease, Department of Targeted Intervention, Division of Surgery and Interventional Science, University College London, Charles Bell House, 43-45 Foley Street, London W1W 7TS, UK
| |
Collapse
|
7
|
Micalet A, Tappouni LJ, Peszko K, Karagianni D, Lam A, Counsell JR, Quezada SA, Moeendarbary E, Cheema U. Urokinase-type plasminogen activator (uPA) regulates invasion and matrix remodelling in colorectal cancer. Matrix Biol Plus 2023; 19-20:100137. [PMID: 38020586 PMCID: PMC10667746 DOI: 10.1016/j.mbplus.2023.100137] [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: 07/24/2023] [Revised: 11/02/2023] [Accepted: 11/13/2023] [Indexed: 12/01/2023] Open
Abstract
Background Cancer cells remodel their local physical environment through processes of matrix reorganisation, deposition, stiffening and degradation. Urokinase-type plasminogen activator (uPA), which is encoded by the PLAU gene, is an extracellular proteolytic enzyme known to be involved in cancer progression and tumour microenvironment (TME) remodelling. Perturbing uPA therefore has a strong potential as a mechano-based cancer therapy. This work is a bioengineering investigation to validate whether 1) uPA is involved in matrix degradation and 2) preventing matrix degradation by targeting uPA can reduce cancer cell invasion and metastasis. Methods To this aim, we used an engineered 3D in vitro model, termed the tumouroid, that appropriately mimics the tumour's native biophysical environment (3 kPa). A CRISPR-Cas9 mediated uPA knockout was performed to introduce a loss of function mutation in the gene coding sequence. Subsequently, to validate the translational potential of blocking uPA action, we tested a pharmacological inhibitor, UK-371,801. The changes in matrix stiffness were measured by atomic force microscopy (AFM). Invasion was quantified using images of the tumouroid, obtained after 21 days of culture. Results We showed that uPA is highly expressed in invasive breast and colorectal cancers, and these invasive cancer cells locally degrade their TME. PLAU (uPA) gene knock-out (KO) completely stopped matrix remodelling and significantly reduced cancer invasion. Many invasive cancer gene markers were also downregulated in the PLAU KO tumouroids. Pharmacological inhibition of uPA showed similarly promising results, where matrix degradation was reduced and so was the cancer invasion. Conclusion This work supports the role of uPA in matrix degradation. It demonstrates that the invasion of cancer cells was significantly reduced when enzymatic breakdown of the TME matrix was prevented. Collectively, this provides strong evidence of the effectiveness of targeting uPA as a mechano-based cancer therapy.
Collapse
Affiliation(s)
- Auxtine Micalet
- UCL Centre for 3D Models of Health and Disease, Department of Targeted Intervention, Division of Surgery and Interventional Science, University College London, Charles Bell House, 43-45 Foley Street, London W1W 7TS, United Kingdom
- Department of Mechanical Engineering, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - Luke J. Tappouni
- UCL Centre for Targeted Cancer Therapies, Department of Targeted Intervention, Division of Surgery and Interventional Science, University College London, Charles Bell House, 43-45 Foley Street, London W1W 7TS, United Kingdom
| | - Katarzyna Peszko
- UCL Centre for 3D Models of Health and Disease, Department of Targeted Intervention, Division of Surgery and Interventional Science, University College London, Charles Bell House, 43-45 Foley Street, London W1W 7TS, United Kingdom
| | - Despoina Karagianni
- Immune Regulation and Tumour Immunotherapy Group, UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, United Kingdom
| | - Ashley Lam
- UCL Centre for 3D Models of Health and Disease, Department of Targeted Intervention, Division of Surgery and Interventional Science, University College London, Charles Bell House, 43-45 Foley Street, London W1W 7TS, United Kingdom
| | - John R. Counsell
- UCL Centre for Targeted Cancer Therapies, Department of Targeted Intervention, Division of Surgery and Interventional Science, University College London, Charles Bell House, 43-45 Foley Street, London W1W 7TS, United Kingdom
| | - Sergio A. Quezada
- Immune Regulation and Tumour Immunotherapy Group, UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, United Kingdom
| | - Emad Moeendarbary
- Department of Mechanical Engineering, University College London, Gower Street, London WC1E 6BT, United Kingdom
- 199 Biotechnologies Ltd., Gloucester Road, London W2 6LD, United Kingdom
| | - Umber Cheema
- UCL Centre for 3D Models of Health and Disease, Department of Targeted Intervention, Division of Surgery and Interventional Science, University College London, Charles Bell House, 43-45 Foley Street, London W1W 7TS, United Kingdom
| |
Collapse
|
8
|
Schab AM, Greenwade MM, Stock E, Lomonosova E, Cho K, Grither WR, Noia H, Wilke D, Mullen MM, Hagemann AR, Hagemann IS, Thaker PH, Kuroki LM, McCourt CK, Khabele D, Powell MA, Mutch DG, Zhao P, Shriver LP, Patti GJ, Longmore GD, Fuh KC. Stromal DDR2 Promotes Ovarian Cancer Metastasis through Regulation of Metabolism and Secretion of Extracellular Matrix Proteins. Mol Cancer Res 2023; 21:1234-1248. [PMID: 37527178 PMCID: PMC10832402 DOI: 10.1158/1541-7786.mcr-23-0347] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/03/2023] [Accepted: 07/28/2023] [Indexed: 08/03/2023]
Abstract
Ovarian cancer is the leading cause of gynecologic cancer-related deaths. The propensity for metastasis within the peritoneal cavity is a driving factor for the poor outcomes associated with this disease, but there is currently no effective therapy targeting metastasis. In this study, we investigate the contribution of stromal cells to ovarian cancer metastasis and identify normal stromal cell expression of the collagen receptor, discoidin domain receptor 2 (DDR2), that acts to facilitate ovarian cancer metastasis. In vivo, global genetic inactivation of Ddr2 impairs the ability of Ddr2-expressing syngeneic ovarian cancer cells to spread throughout the peritoneal cavity. Specifically, DDR2 expression in mesothelial cells lining the peritoneal cavity facilitates tumor cell attachment and clearance. Subsequently, omentum fibroblast expression of DDR2 promotes tumor cell invasion. Mechanistically, we find DDR2-expressing fibroblasts are more energetically active, such that DDR2 regulates glycolysis through AKT/SNAI1 leading to suppressed fructose-1,6-bisphosphatase and increased hexokinase activity, a key glycolytic enzyme. Upon inhibition of DDR2, we find decreased protein synthesis and secretion. Consequently, when DDR2 is inhibited, there is reduction in secreted extracellular matrix proteins important for metastasis. Specifically, we find that fibroblast DDR2 inhibition leads to decreased secretion of the collagen crosslinker, LOXL2. Adding back LOXL2 to DDR2 deficient fibroblasts rescues the ability of tumor cells to invade. Overall, our results suggest that stromal cell expression of DDR2 is an important mediator of ovarian cancer metastasis. IMPLICATIONS DDR2 is highly expressed by stromal cells in ovarian cancer that can mediate metastasis and is a potential therapeutic target in ovarian cancer.
Collapse
Affiliation(s)
- Angela M. Schab
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
- Center for Reproductive Health Sciences, Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
| | - Molly M. Greenwade
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
- Center for Reproductive Health Sciences, Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
| | - Elizabeth Stock
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
- Center for Reproductive Health Sciences, Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
| | - Elena Lomonosova
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
- Center for Reproductive Health Sciences, Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
| | - Kevin Cho
- Center for Metabolomics and Isotope Tracing, Department of Chemistry, Department of Medicine, Washington University, St. Louis, MO 63110, USA
| | - Whitney R. Grither
- Department of Obstetrics and Gynecology, Barnes Jewish Hospital, Washington University, St. Louis, MO 63110, USA
| | - Hollie Noia
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
- Center for Reproductive Health Sciences, Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
| | - Daniel Wilke
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
- Center for Reproductive Health Sciences, Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
| | - Mary M. Mullen
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
- Center for Reproductive Health Sciences, Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
| | - Andrea R. Hagemann
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
| | - Ian S. Hagemann
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
| | - Premal H. Thaker
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
| | - Lindsay M. Kuroki
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
| | - Carolyn K. McCourt
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
| | - Dineo Khabele
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
- Center for Reproductive Health Sciences, Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
| | - Matthew A. Powell
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
| | - David G. Mutch
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
| | - Peinan Zhao
- Department of Obstetrics and Gynecology, Barnes Jewish Hospital, Washington University, St. Louis, MO 63110, USA
| | - Leah P. Shriver
- Center for Metabolomics and Isotope Tracing, Department of Chemistry, Department of Medicine, Washington University, St. Louis, MO 63110, USA
| | - Gary J. Patti
- Center for Metabolomics and Isotope Tracing, Department of Chemistry, Department of Medicine, Washington University, St. Louis, MO 63110, USA
| | - Gregory D. Longmore
- Division of Oncology, Department of Medicine Washington University, St. Louis. MO 63110, USA
- ICCE Institute, Washington University, St. Louis MO 63110, USA
| | - Katherine C. Fuh
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
- Center for Reproductive Health Sciences, Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology University of California, San Francisco, San Francisco, CA 94143 USA
| |
Collapse
|
9
|
Löser R, Kuchar M, Wodtke R, Neuber C, Belter B, Kopka K, Santhanam L, Pietzsch J. Lysyl Oxidases as Targets for Cancer Therapy and Diagnostic Imaging. ChemMedChem 2023; 18:e202300331. [PMID: 37565736 DOI: 10.1002/cmdc.202300331] [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: 06/28/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 08/12/2023]
Abstract
The understanding of the contribution of the tumour microenvironment to cancer progression and metastasis, in particular the interplay between tumour cells, fibroblasts and the extracellular matrix has grown tremendously over the last years. Lysyl oxidases are increasingly recognised as key players in this context, in addition to their function as drivers of fibrotic diseases. These insights have considerably stimulated drug discovery efforts towards lysyl oxidases as targets over the last decade. This review article summarises the biochemical and structural properties of theses enzymes. Their involvement in tumour progression and metastasis is highlighted from a biochemical point of view, taking into consideration both the extracellular and intracellular action of lysyl oxidases. More recently reported inhibitor compounds are discussed with an emphasis on their discovery, structure-activity relationships and the results of their biological characterisation. Molecular probes developed for imaging of lysyl oxidase activity are reviewed from the perspective of their detection principles, performance and biomedical applications.
Collapse
Affiliation(s)
- Reik Löser
- Institute of Radiopharmaceutical Cancer Research Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Mommsenstraße 4, 01069, Dresden, Germany
| | - Manuela Kuchar
- Institute of Radiopharmaceutical Cancer Research Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Robert Wodtke
- Institute of Radiopharmaceutical Cancer Research Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Christin Neuber
- Institute of Radiopharmaceutical Cancer Research Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Birgit Belter
- Institute of Radiopharmaceutical Cancer Research Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Klaus Kopka
- Institute of Radiopharmaceutical Cancer Research Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Mommsenstraße 4, 01069, Dresden, Germany
| | - Lakshmi Santhanam
- Departments of Anesthesiology and Critical Care Medicine and Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Jens Pietzsch
- Institute of Radiopharmaceutical Cancer Research Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Mommsenstraße 4, 01069, Dresden, Germany
| |
Collapse
|
10
|
Behnam B, Taghizadeh-Hesary F. Mitochondrial Metabolism: A New Dimension of Personalized Oncology. Cancers (Basel) 2023; 15:4058. [PMID: 37627086 PMCID: PMC10452105 DOI: 10.3390/cancers15164058] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Energy is needed by cancer cells to stay alive and communicate with their surroundings. The primary organelles for cellular metabolism and energy synthesis are mitochondria. Researchers recently proved that cancer cells can steal immune cells' mitochondria using nanoscale tubes. This finding demonstrates the dependence of cancer cells on normal cells for their living and function. It also denotes the importance of mitochondria in cancer cells' biology. Emerging evidence has demonstrated how mitochondria are essential for cancer cells to survive in the harsh tumor microenvironments, evade the immune system, obtain more aggressive features, and resist treatments. For instance, functional mitochondria can improve cancer resistance against radiotherapy by scavenging the released reactive oxygen species. Therefore, targeting mitochondria can potentially enhance oncological outcomes, according to this notion. The tumors' responses to anticancer treatments vary, ranging from a complete response to even cancer progression during treatment. Therefore, personalized cancer treatment is of crucial importance. So far, personalized cancer treatment has been based on genomic analysis. Evidence shows that tumors with high mitochondrial content are more resistant to treatment. This paper illustrates how mitochondrial metabolism can participate in cancer resistance to chemotherapy, immunotherapy, and radiotherapy. Pretreatment evaluation of mitochondrial metabolism can provide additional information to genomic analysis and can help to improve personalized oncological treatments. This article outlines the importance of mitochondrial metabolism in cancer biology and personalized treatments.
Collapse
Affiliation(s)
- Babak Behnam
- Department of Regulatory Affairs, Amarex Clinical Research, NSF International, Germantown, MD 20874, USA
| | - Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran 1445613131, Iran
- Department of Radiation Oncology, Iran University of Medical Sciences, Tehran 1445613131, Iran
| |
Collapse
|
11
|
Zhu S, Zhang T, Gao H, Jin G, Yang J, He X, Guo H, Xu F. Combination Therapy of Lox Inhibitor and Stimuli-Responsive Drug for Mechanochemically Synergistic Breast Cancer Treatment. Adv Healthc Mater 2023; 12:e2300103. [PMID: 37099721 DOI: 10.1002/adhm.202300103] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 04/21/2023] [Indexed: 04/28/2023]
Abstract
Chemotherapy based on small molecule drugs, hormones, cycline kinase inhibitors, and monoclonal antibodies has been widely used for breast cancer treatment in the clinic but with limited efficacy, due to the poor specificity and tumor microenvironment (TME)-caused diffusion barrier. Although monotherapies targeting biochemical cues or physical cues in the TME have been developed, none of them can cope with the complex TME, while mechanochemical combination therapy remains largely to be explored. Herein, a combination therapy strategy based on an extracellular matrix (ECM) modulator and TME-responsive drug for the first attempt of mechanochemically synergistic treatment of breast cancer is developed. Specifically, based on overexpressed NAD(P)H quinone oxidoreductase 1 (NQO1) in breast cancer, a TME-responsive drug (NQO1-SN38) is designed and it is combined with the inhibitor (i.e., β-Aminopropionitrile, BAPN) for Lysyl oxidases (Lox) that contributes to the tumor stiffness, for mechanochemical therapy. It is demonstrated that NQO1 can trigger the degradation of NQO1-SN38 and release SN38, showing nearly twice tumor inhibition efficiency compared with SN38 treatment in vitro. Lox inhibition with BAPN significantly reduces collagen deposition and enhances drug penetration in tumor heterospheroids in vitro. It is further demonstrated that the mechanochemical therapy showed outstanding therapeutic efficacy in vivo, providing a promising approach for breast cancer therapy.
Collapse
Affiliation(s)
- Shanshan Zhu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering & Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Tian Zhang
- Bioinspired Engineering & Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Huan Gao
- Bioinspired Engineering & Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Guorui Jin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering & Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Jin Yang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Xiaocong He
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering & Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Hui Guo
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering & Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| |
Collapse
|
12
|
Zhao J, Jia X, Li Q, Zhang H, Wang J, Huang S, Hu Z, Li C. Genomic and transcriptional characterization of early esophageal squamous cell carcinoma. BMC Med Genomics 2023; 16:153. [PMID: 37393256 PMCID: PMC10315050 DOI: 10.1186/s12920-023-01588-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/20/2023] [Indexed: 07/03/2023] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a highly heterogeneous cancer that lacks comprehensive understanding and effective treatment. Although multi-omics study has revealed features and underlying drivers of advanced ESCC, research on molecular characteristics of the early stage ESCC is quite limited. MATERIALS AND METHODS We presented characteristics of genomics and transcriptomics in 10 matched pairs of tumor and normal tissues of early ESCC patients in the China region. RESULTS We identified the specific patterns of cancer gene mutations and copy number variations. We also found a dramatic change in the transcriptome, with more than 4,000 genes upregulated in cancer. Among them, more than one-third of HOX family genes were specifically and highly expressed in early ESCC samples of China and validated by RT-qPCR. Gene regulation network analysis indicated that alteration of Hox family genes promoted the proliferation and metabolism remodeling of early ESCC. CONCLUSIONS We characterized the genomic and transcriptomic landscape of 10 paired normal adjacent and early ESCC tissues in the China region, and provided a new perspective to understand the development of ESCC and insight into potential prevention and diagnostic targets for the management of early ESCC in China.
Collapse
Affiliation(s)
- Jingjing Zhao
- Department of Gastroenterology, Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xiya Jia
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Qiaojuan Li
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Hena Zhang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jianjun Wang
- Department of Pediatric Medicine, Gansu Provincial People's Hospital, Lanzhou City, , Gansu Province, China
| | - Shenglin Huang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zhixiang Hu
- Department of Gastroenterology, Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Caiping Li
- Department of Gastroenterology, Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.
| |
Collapse
|
13
|
Bui CB, To KD, Vu DM, Nguyen QG, Nguyen HT, Nguyen SB. Denatured collagen inhibits neuroblastoma tumor-sphere migration and growth via the LOX/LOXL2 - FAK signaling pathway. J Therm Biol 2023; 115:103624. [PMID: 37399743 DOI: 10.1016/j.jtherbio.2023.103624] [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: 06/13/2022] [Revised: 04/16/2023] [Accepted: 06/11/2023] [Indexed: 07/05/2023]
Abstract
A complex interplay exists within the tumor microenvironment and extracellular matrix, which could contribute to solid tumor progression. Collagen, a major component of the extracellular matrix, may correlate with cancer prognosis. While thermal ablation has shown promise as a minimally invasive treatment of solid tumors, its impact on collagen is still unknown. In this study, we demonstrate that thermal ablation, but not cryo-ablation, induces irreversible collagen denaturation in a neuroblastoma sphere model. Prolonged collagen denaturation resulted in a significant reduction in sphere stiffness, migration, and proliferation, and an increase in apoptosis. Mechanistic analysis revealed that collagen denaturation inhibited collagen cross-linking, reduced extracellular LOX/LOXL2 expression, and resulted in decreased phosphorylation of FAK. Downstream of FAK, we observed reduced epithelial to mesenchymal transition, attenuated CDC42 expression, and decreased migration. Collectively, these results suggest that denatured collagen presents a novel target for modulating the tumor microenvironment and treating solid cancers via the LOX1/LOXL2-FAK signaling pathway.
Collapse
Affiliation(s)
- Chi-Bao Bui
- Unit of Molecular Biology, City Children's Hospital, Ho Chi Minh City, Vietnam; School of Medicine, Ho Chi Minh City, Vietnam; Vietnam National University, Ho Chi Minh City, Vietnam
| | - Kha Dong To
- School of Medicine, Ho Chi Minh City, Vietnam; University College London, London, United Kingdom; Vietnam National University, Ho Chi Minh City, Vietnam
| | - Diem My Vu
- Center for Molecular Biomedicine, University of Medicine and Pharmacy at Ho Chi Minh City, Vietnam
| | - Quynh-Giang Nguyen
- School of Medicine, Ho Chi Minh City, Vietnam; Vietnam National University, Ho Chi Minh City, Vietnam
| | - Hiep Thi Nguyen
- School of Biomedical Engineering, International University, Ho Chi Minh City, Vietnam
| | - Si-Bao Nguyen
- School of Medicine, Ho Chi Minh City, Vietnam; Vietnam National University, Ho Chi Minh City, Vietnam.
| |
Collapse
|
14
|
Bernabei I, Hansen U, Ehirchiou D, Brinckmann J, Chobaz V, Busso N, Nasi S. CD11b Deficiency Favors Cartilage Calcification via Increased Matrix Vesicles, Apoptosis, and Lysyl Oxidase Activity. Int J Mol Sci 2023; 24:ijms24119776. [PMID: 37298730 DOI: 10.3390/ijms24119776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/31/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Pathological cartilage calcification is a hallmark feature of osteoarthritis, a common degenerative joint disease, characterized by cartilage damage, progressively causing pain and loss of movement. The integrin subunit CD11b was shown to play a protective role against cartilage calcification in a mouse model of surgery-induced OA. Here, we investigated the possible mechanism by which CD11b deficiency could favor cartilage calcification by using naïve mice. First, we found by transmission electron microscopy (TEM) that CD11b KO cartilage from young mice presented early calcification spots compared with WT. CD11b KO cartilage from old mice showed progression of calcification areas. Mechanistically, we found more calcification-competent matrix vesicles and more apoptosis in both cartilage and chondrocytes isolated from CD11b-deficient mice. Additionally, the extracellular matrix from cartilage lacking the integrin was dysregulated with increased collagen fibrils with smaller diameters. Moreover, we revealed by TEM that CD11b KO cartilage had increased expression of lysyl oxidase (LOX), the enzyme that catalyzes matrix crosslinks. We confirmed this in murine primary CD11b KO chondrocytes, where Lox gene expression and crosslinking activity were increased. Overall, our results suggest that CD11b integrin regulates cartilage calcification through reduced MV release, apoptosis, LOX activity, and matrix crosslinking. As such, CD11b activation might be a key pathway for maintaining cartilage integrity.
Collapse
Affiliation(s)
- Ilaria Bernabei
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Uwe Hansen
- Institute for Musculoskeletal Medicine, University Hospital of Münster, 48149 Münster, Germany
| | - Driss Ehirchiou
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Jürgen Brinckmann
- Department of Dermatology, University of Lübeck, 23562 Lübeck, Germany
| | - Veronique Chobaz
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Nathalie Busso
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Sonia Nasi
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, 1011 Lausanne, Switzerland
| |
Collapse
|
15
|
Micalet A, Pape J, Bakkalci D, Javanmardi Y, Hall C, Cheema U, Moeendarbary E. Evaluating the Impact of a Biomimetic Mechanical Environment on Cancer Invasion and Matrix Remodeling. Adv Healthc Mater 2023; 12:e2201749. [PMID: 36333907 PMCID: PMC11468596 DOI: 10.1002/adhm.202201749] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/10/2022] [Indexed: 10/13/2024]
Abstract
The stiffness of tumors and their host tissues is much higher than most hydrogels, which are conventionally used to study in vitro cancer progression. The tumoroid assay is an engineered 3D in vitro tumor model that allows investigation of cancer cell invasion in an environment that is biomimetic in terms of extracellular matrix (ECM) composition and stiffness. Using this model, the change in matrix stiffness by epithelial colorectal cancer cells is systematically characterized by atomic force microscopy indentation tests. Less invasive epithelial cancer cells stiffen the tumor microenvironment while highly aggressive epithelial cancer cells show significant softening of the tumor microenvironment. Changes in stiffness are attributed to both cell-generated active forces as well as ECM degradation and remodeling. The degradation is in part attributed to the enzymatic activity of matrix metalloproteinases (MMPs) as demonstrated by the significant expression of MMP-2 and MMP-9 at both gene and protein levels. Targeting MMP activity through broad-spectrum drug inhibition (BB-94) reverses the changes in stiffness and also decreases cancer cell invasion. These results promote the idea of using mechano-based cancer therapies such as MMP inhibition.
Collapse
Affiliation(s)
- Auxtine Micalet
- Department of Mechanical EngineeringUniversity College LondonGower StreetLondonWC1E 6BTUK
- UCL Centre for 3D Models of Health and DiseaseDepartment of Targeted InterventionDivision of Surgery and Interventional ScienceUniversity College LondonCharles Bell House43–45 Foley StreetLondonW1W 7TSUK
| | - Judith Pape
- UCL Centre for 3D Models of Health and DiseaseDepartment of Targeted InterventionDivision of Surgery and Interventional ScienceUniversity College LondonCharles Bell House43–45 Foley StreetLondonW1W 7TSUK
| | - Deniz Bakkalci
- UCL Centre for 3D Models of Health and DiseaseDepartment of Targeted InterventionDivision of Surgery and Interventional ScienceUniversity College LondonCharles Bell House43–45 Foley StreetLondonW1W 7TSUK
| | - Yousef Javanmardi
- Department of Mechanical EngineeringUniversity College LondonGower StreetLondonWC1E 6BTUK
| | - Chloe Hall
- Department of Mechanical EngineeringUniversity College LondonGower StreetLondonWC1E 6BTUK
| | - Umber Cheema
- UCL Centre for 3D Models of Health and DiseaseDepartment of Targeted InterventionDivision of Surgery and Interventional ScienceUniversity College LondonCharles Bell House43–45 Foley StreetLondonW1W 7TSUK
| | - Emad Moeendarbary
- Department of Mechanical EngineeringUniversity College LondonGower StreetLondonWC1E 6BTUK
| |
Collapse
|
16
|
Melamed S, Zaffryar-Eilot S, Nadjar-Boger E, Aviram R, Zhao H, Yaseen-Badarne W, Kalev-Altman R, Sela-Donenfeld D, Lewinson O, Astrof S, Hasson P, Wolfenson H. Initiation of fibronectin fibrillogenesis is an enzyme-dependent process. Cell Rep 2023; 42:112473. [PMID: 37148241 DOI: 10.1016/j.celrep.2023.112473] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/16/2023] [Accepted: 04/19/2023] [Indexed: 05/08/2023] Open
Abstract
Fibronectin fibrillogenesis and mechanosensing both depend on integrin-mediated force transmission to the extracellular matrix. However, force transmission is in itself dependent on fibrillogenesis, and fibronectin fibrils are found in soft embryos where high forces cannot be applied, suggesting that force cannot be the sole initiator of fibrillogenesis. Here, we identify a nucleation step prior to force transmission, driven by fibronectin oxidation mediated by lysyl oxidase enzyme family members. This oxidation induces fibronectin clustering, which promotes early adhesion, alters cellular response to soft matrices, and enhances force transmission to the matrix. In contrast, absence of fibronectin oxidation abrogates fibrillogenesis, perturbs cell-matrix adhesion, and compromises mechanosensation. Moreover, fibronectin oxidation promotes cancer cell colony formation in soft agar as well as collective and single-cell migration. These results reveal a force-independent enzyme-dependent mechanism that initiates fibronectin fibrillogenesis, establishing a critical step in cell adhesion and mechanosensing.
Collapse
Affiliation(s)
- Shay Melamed
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Shelly Zaffryar-Eilot
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Elisabeth Nadjar-Boger
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Rohtem Aviram
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Huaning Zhao
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ 07103, USA
| | - Wesal Yaseen-Badarne
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Rotem Kalev-Altman
- Koret School of Veterinary Medicine, Faculty of Agriculture, Food and Environmental Sciences, The Hebrew University, Rehovot, Israel
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, Faculty of Agriculture, Food and Environmental Sciences, The Hebrew University, Rehovot, Israel
| | - Oded Lewinson
- Department of Molecular Microbiology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Sophie Astrof
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ 07103, USA
| | - Peleg Hasson
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel.
| | - Haguy Wolfenson
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel.
| |
Collapse
|
17
|
Sharma RK, Kamble SH, Krishnan S, Gomes J, To B, Li S, Liu IC, Gumz ML, Mohandas R. Involvement of lysyl oxidase in the pathogenesis of arterial stiffness in chronic kidney disease. Am J Physiol Renal Physiol 2023; 324:F364-F373. [PMID: 36825626 PMCID: PMC10069822 DOI: 10.1152/ajprenal.00239.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 02/01/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Patients with chronic kidney disease (CKD) are at increased risk for adverse cardiovascular events. CKD is associated with increases in arterial stiffness, whereas improvements in arterial stiffness correlate with better survival. However, arterial stiffness is increased early in CKD, suggesting that there might be additional factors, unique to kidney disease, that increase arterial stiffness. Lysyl oxidase (LOX) is a key mediator of collagen cross linking and matrix remodeling. LOX is predominantly expressed in the cardiovascular system, and its upregulation has been associated with increased tissue stiffening and extracellular matrix remodeling. Thus, this study was designed to evaluate the role of increased LOX activity in inducing aortic stiffness in CKD and whether β-aminopropionitrile (BAPN), a LOX inhibitor, could prevent aortic stiffness by reducing collagen cross linking. Eight-week-old male C57BL/6 mice were subjected to 5/6 nephrectomy (Nx) or sham surgery. Two weeks after surgery, mice were randomized to BAPN (300 mg/kg/day in water) or vehicle treatment for 4 wk. Aortic stiffness was assessed by pulse wave velocity (PWV) using Doppler ultrasound. Aortic levels of LOX were assessed by ELISA, and cross-linked total collagen levels were analyzed by mass spectrometry and Sircol assay. Nx mice showed increased PWV and aortic wall remodeling compared with control mice. Collagen cross linking was increased in parallel with the increases in total collagen in the aorta of Nx mice. In contrast, Nx mice that received BAPN treatment showed decreased cross-linked collagens and PWV compared with that received vehicle treatment. Our results indicated that LOX might be an early and key mediator of aortic stiffness in CKD.NEW & NOTEWORTHY Arterial stiffness in CKD is associated with adverse cardiovascular outcomes. However, the mechanisms underlying increased aortic stiffness in CKD are unclear. Herein, we demonstrated that 1) increased aortic stiffness in CKD is independent of hypertension and calcification and 2) LOX-mediated changes in extracellular matrix are at least in part responsible for increased aortic stiffness in CKD. Prevention of excess LOX may have therapeutic potential in alleviating increased aortic stiffness and improving cardiovascular disease in CKD.
Collapse
Affiliation(s)
- Ravindra K Sharma
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, United States
| | - Shyam H Kamble
- Department of Pharmacology, University of Florida College of Medicine, Gainesville, Florida, United States
| | - Suraj Krishnan
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, United States
| | - Joshua Gomes
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, United States
| | - Brandon To
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, United States
| | - Shiyu Li
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, United States
| | - I-Chia Liu
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, United States
| | - Michelle L Gumz
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, Florida, United States
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, Florida, United States
| | - Rajesh Mohandas
- Division of Nephrology and Hypertension, Louisiana State University Health Sciences Center School of Medicine, New Orleans, Louisiana, United States
| |
Collapse
|
18
|
Coffelt SB, Morton JP. LOXL2 in pancreatic tumourigenesis: the complexity of tumour-stromal crosstalk exemplified. Gut 2023; 72:221-222. [PMID: 35428658 DOI: 10.1136/gutjnl-2022-327430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 04/07/2022] [Indexed: 01/10/2023]
Affiliation(s)
- Seth B Coffelt
- Cancer Research UK Beatson Institute, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Jennifer P Morton
- Cancer Research UK Beatson Institute, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
19
|
Zulaziz N, Chai SJ, Lim KP. The origins, roles and therapies of cancer associated fibroblast in liver cancer. Front Oncol 2023; 13:1151373. [PMID: 37035187 PMCID: PMC10076538 DOI: 10.3389/fonc.2023.1151373] [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: 01/26/2023] [Accepted: 03/03/2023] [Indexed: 04/11/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common form of liver cancer. It is often preceded by chronic inflammation such as liver fibrosis and cirrhosis. Different cell types are believed to give rise to liver-specific cancer associated fibroblast (CAF), these include resident fibroblast, hepatic stellate cell, liver cancer cell, hepatic sinusoidal endothelial cell and mesenchymal stromal cell. The abundance of fibroblasts has contributed to the cancer progression, immune modulation and treatment resistance in HCC. In this review, we discussed the origins, subtypes and roles of cancer associated fibroblasts in HCC. Their specific roles in shaping the tumor microenvironment, facilitating cancer growth, and modulating different immune cell types to confer a permissive environment for cancer growth. CAF is now an attractive therapeutic target for cancer treatment, however specific therapeutic development in HCC is still lacking. Hence, we have included preclinical and clinical development of CAF-specific interventions for other cancer types in this review. However, most CAF-specific therapies have resulted in disappointing clinical outcomes, likely due to the difficulties in differentiating CAF from normal fibroblast. A thorough understanding of the characteristics and functionalities of CAF is warranted to further improve the therapeutic efficacy of anti-CAF therapies.
Collapse
|
20
|
Komalasari NLGY, Tomonobu N, Kinoshita R, Chen Y, Sakaguchi Y, Gohara Y, Jiang F, Yamamoto KI, Murata H, Ruma IMW, Sumardika IW, Zhou J, Yamauchi A, Kuribayashi F, Inoue Y, Toyooka S, Sakaguchi M. Lysyl oxidase-like 4 exerts an atypical role in breast cancer progression that is dependent on the enzymatic activity that targets the cell-surface annexin A2. Front Oncol 2023; 13:1142907. [PMID: 37091157 PMCID: PMC10114587 DOI: 10.3389/fonc.2023.1142907] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/13/2023] [Indexed: 04/25/2023] Open
Abstract
Background LOX family members are reported to play pivotal roles in cancer. Unlike their enzymatic activities in collagen cross-linking, their precise cancer functions are unclear. We revealed that LOXL4 is highly upregulated in breast cancer cells, and we thus sought to define an unidentified role of LOXL4 in breast cancer. Methods We established the MDA-MB-231 sublines MDA-MB-231-LOXL4 mutCA and -LOXL4 KO, which stably overexpress mutant LOXL4 that loses its catalytic activity and genetically ablates the intrinsic LOXL4 gene, respectively. In vitro and in vivo evaluations of these cells' activities of cancer outgrowth were conducted by cell-based assays in cultures and an orthotopic xenograft model, respectively. The new target (s) of LOXL4 were explored by the MS/MS analytic approach. Results Our in vitro results revealed that both the overexpression of mutCA and the KO of LOXL4 in cells resulted in a marked reduction of cell growth and invasion. Interestingly, the lowered cellular activities observed in the engineered cells were also reflected in the mouse model. We identified a novel binding partner of LOXL4, i.e., annexin A2. LOXL4 catalyzes cell surface annexin A2 to achieve a cross-linked multimerization of annexin A2, which in turn prevents the internalization of integrin β-1, resulting in the locking of integrin β-1 on the cell surface. These events enhance the promotion of cancer cell outgrowth. Conclusions LOXL4 has a new role in breast cancer progression that occurs via an interaction with annexin A2 and integrin β-1 on the cell surface.
Collapse
Affiliation(s)
- Ni Luh Gede Yoni Komalasari
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Okayama, Japan
- Faculty of Medicine, Udayana University, Denpasar, Bali, Indonesia
| | - Nahoko Tomonobu
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Rie Kinoshita
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Youyi Chen
- Department of General Surgery & Bio-Bank of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yoshihiko Sakaguchi
- Department of Microbiology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Yuma Gohara
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Fan Jiang
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Ken-ich Yamamoto
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Hitoshi Murata
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | | | | | - Jin Zhou
- Medical Oncology Department of Gastrointestinal Tumors, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning, China
| | - Akira Yamauchi
- Department of Biochemistry, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Futoshi Kuribayashi
- Department of Biochemistry, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Yusuke Inoue
- Faculty of Science and Technology, Division of Molecular Science, Gunma University, Kiryu, Gunma, Japan
| | - Shinichi Toyooka
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Masakiyo Sakaguchi
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Okayama, Japan
- *Correspondence: Masakiyo Sakaguchi,
| |
Collapse
|
21
|
Zhang X, Wu X, Sun Y, Chu Y, Liu F, Chen C. TRIM44 regulates tumor immunity in gastric cancer through LOXL2-dependent extracellular matrix remodeling. Cell Oncol (Dordr) 2022; 46:423-435. [PMID: 36512309 DOI: 10.1007/s13402-022-00759-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 11/12/2022] [Accepted: 12/06/2022] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Gastric cancer is a gastrointestinal malignancy with high mortality and poor prognosis, and the molecular mechanism of gastric tumorigenesis remains unclear. TRIM44 has been reported to be involved in tumor development. However, the role of TRIM44 in tumor immunity is largely unknown. METHODS We analyzed TRIM44 expression in clinical gastric cancer tissues and normal tissues by using western blot, quantitative real-time PCR and bioinformatics analyses. We further investigated the involvement of TRIM44 in tumor immunity in vivo and found that it was dependent on extracellular matrix remodeling. We detected the interaction between TRIM44 and LOXL2 by using immunofluorescence staining and coimmunoprecipitation assays. We observed that TRIM44 mediates the stability of LOXL2 by ubiquitination assays. RESULTS TRIM44 expression is high and is correlated with T-cell infiltration in gastric cancer. TRIM44 inhibits gastric tumorigenicity by regulating T-cell-mediated antitumor immunity and modulating the protein level of LOXL2. Mechanistically, TRIM44 directly binds to LOXL2 and affects the stability of LOXL2 to change extracellular matrix remodeling and influence tumor immunity. CONCLUSION These findings demonstrate that TRIM44 regulates the stability of LOXL2 to remodel the tumor extracellular matrix to modulate tumor immunity in gastric cancer and that the TRIM44/LOXL2 complex is a promising biomarker for gastric cancer prognosis and might be a novel immunotherapy target.
Collapse
Affiliation(s)
- Xin Zhang
- Department of General Surgery, Qilu Hospital of Shandong University, 107 West Wenhua Road, 250012, JiNan, China
| | - Xiusheng Wu
- Department of General Surgery, Linyi People's Hospital, 105 Plaza Street, Linyi County, China
| | - Ying Sun
- Department of Blood quality Control, Yantai central blood station, 10 Haiyun Road, Yantai, China
| | - Yali Chu
- Department of General Surgery, Qilu Hospital of Shandong University, 107 West Wenhua Road, 250012, JiNan, China
| | - Fengjun Liu
- Department of General Surgery, Qilu Hospital of Shandong University, 107 West Wenhua Road, 250012, JiNan, China
| | - Cheng Chen
- Department of General Surgery, Qilu Hospital of Shandong University, 107 West Wenhua Road, 250012, JiNan, China.
| |
Collapse
|
22
|
Prognosis Risk Model Based on Necroptosis-Related Signature for Bladder Cancer. Genes (Basel) 2022; 13:genes13112120. [DOI: 10.3390/genes13112120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/13/2022] [Accepted: 11/10/2022] [Indexed: 11/18/2022] Open
Abstract
Background: Bladder cancer(BLCA) is the ninth most common cancer. In recent years, necroptosis was found to be related to the occurrence and development of tumors. In this study, we aimed to construct a model based on a necroptosis-related signature to evaluate the potential prognostic application in BLCA. Methods: A total of 67 necroptosis-related genes were used to select the ideal cluster numbers, and it was found that there were four necroptosis-related patterns. Then, we compared the gene expression levels among all of the groups and established a necroptosis-related prognostic model. We made the following enrichment analysis of function and built a novel scoring system, the NEC score, to evaluate the state of necroptosis according to the expression level of necroptosis-related genes. Results: A total of 67 necroptosis-related genes were used to define four distinct necroptosis-related patterns: NEC cluster1–4. Each NEC cluster exhibited different patterns of survival and immune infiltration. Based on univariate Cox regression analyses and least absolute shrinkage and selection operator (Lasso) regression, 14 necroptosis-related genes were established to develop the NEC score. Patients were divided into two groups based on the NEC score. Patients in the high NEC score group had a significantly poorer overall survival than those in the low NEC score group. We further confirmed the correlation of clinical characteristics, as well as the immunotherapy outcome, with the NEC score, and confirmed the potential of the NEC score to be an independent prognostic factor. Furthermore, we compared the expression levels of eight potential biomarker genes between our own BLCA tissues and para-carcinoma tissue. Conclusion: We developed a novel NEC score that has a potential prognostic value in BLCA patients and may help personalized immunotherapy counselling.
Collapse
|
23
|
Zhang S, Zhang W, Wu B, Xia L, Li L, Jin K, Zou Y, Sun C. Hub gene target of glioblastoma: LOX, SERPINH1 and TGFBI. Medicine (Baltimore) 2022; 101:e31418. [PMID: 36397358 PMCID: PMC9666166 DOI: 10.1097/md.0000000000031418] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Glioblastoma (GBM) is a malignant tumor. The long-term prognosis of the patients is poor. Therefore, it is of important clinical value to further explore the pathogenesis and look for molecular markers for early diagnosis and targeted treatment. Two expression profiling datasets [GSE50161 (GPL570 platform), GSE116520 (GPL10558 platform)] were respectively downloaded from the gene expression omnibus database. Volcano diagrams show the Differently expressed genes (DEGs) of GSE50161 and GSE116520. A Venn diagram revealed 467 common DEGs between the 2 datasets. Lysyl oxidase (LOX), serpin family H member 1 (SERPINH1) and transforming growth factor beta induced (TGFBI) were negatively correlated with the overall survival rate in patients with GBM. The hub genes are high in GBM tumor tissues. The relative expression levels of LOX, SERPINH1 and TGFBI were significantly higher in GBM samples, compared with the normal brain tissues groups. Bioinformatics technology could be a useful tool to predict progression of GBM and to explore the mechanism of GBM.LOX, SERPINH1 and TGFBI may be involved in the mechanism of the occurrence and development of GBM, and may be used as molecular targets for early diagnosis and specific treatment. DEGs identified using GEO2R. Functional annotation of DEGs using Kyoto Encyclopedia of Genes and Genomes and gene body pathway enrichment analysis. Construction of a protein-protein interaction network. The pathway and process enrichment analysis of the hub genes were performed by Metascape. Survival analysis was performed in gene expression profiling interactive analysis. Real-time fluorescent quantitative polymerase chain reaction assay was performed to verify. The animal model was established for western blot test analysis.
Collapse
Affiliation(s)
- Shuyuan Zhang
- Department of Neurosurgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Weiwei Zhang
- Department of Operating Theater, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Bin Wu
- Department of Neurosurgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Liang Xia
- Department of Neurosurgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Liwen Li
- Department of Neurosurgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Kai Jin
- Department of Neurosurgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Yangfan Zou
- Department of Neurosurgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| | - Caixing Sun
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
- * Correspondence: Caixing Sun, Department of Neurosurgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China (e-mail: )
| |
Collapse
|
24
|
Taghizadeh-Hesary F, Akbari H, Bahadori M, Behnam B. Targeted Anti-Mitochondrial Therapy: The Future of Oncology. Genes (Basel) 2022; 13:1728. [PMID: 36292613 PMCID: PMC9602426 DOI: 10.3390/genes13101728] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/16/2022] [Accepted: 09/22/2022] [Indexed: 11/30/2022] Open
Abstract
Like living organisms, cancer cells require energy to survive and interact with their environment. Mitochondria are the main organelles for energy production and cellular metabolism. Recently, investigators demonstrated that cancer cells can hijack mitochondria from immune cells. This behavior sheds light on a pivotal piece in the cancer puzzle, the dependence on the normal cells. This article illustrates the benefits of new functional mitochondria for cancer cells that urge them to hijack mitochondria. It describes how functional mitochondria help cancer cells' survival in the harsh tumor microenvironment, immune evasion, progression, and treatment resistance. Recent evidence has put forward the pivotal role of mitochondria in the metabolism of cancer stem cells (CSCs), the tumor components responsible for cancer recurrence and metastasis. This theory highlights the mitochondria in cancer biology and explains how targeting mitochondria may improve oncological outcomes.
Collapse
Affiliation(s)
- Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran 1445613131, Iran
- Department of Radiation Oncology, Iran University of Medical Sciences, Tehran 1445613131, Iran
| | - Hassan Akbari
- Department of Pathology, Shahid Beheshti University of Medical Sciences, Tehran P.O. Box 4739-19395, Iran
- Traditional Medicine School, Tehran University of Medical Sciences, Tehran P.O. Box 14155-6559, Iran
| | - Moslem Bahadori
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran P.O. Box 14155-6559, Iran
| | - Babak Behnam
- Department of Regulatory Affairs, Amarex Clinical Research, Germantown, MD 20874, USA
| |
Collapse
|
25
|
Li H, Zu X, Hu J, Xiao Z, Cai Z, Gao N, Chen J. Cuproptosis depicts tumor microenvironment phenotypes and predicts precision immunotherapy and prognosis in bladder carcinoma. Front Immunol 2022; 13:964393. [PMID: 36211344 PMCID: PMC9540537 DOI: 10.3389/fimmu.2022.964393] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Though immune checkpoint inhibitors (ICIs) exhibit durable efficacy in bladder carcinomas (BLCAs), there are still a large portion of patients insensitive to ICIs treatment. METHODS We systematically evaluated the cuproptosis patterns in BLCA patients based on 46 cuproptosis related genes and correlated these cuproptosis patterns with tumor microenvironment (TME) phenotypes and immunotherapy efficacies. Then, for individual patient's evaluation, we constructed a cuproptosis risk score (CRS) for prognosis and a cuproptosis signature for precise TME phenotypes and immunotherapy efficacies predicting. RESULTS Two distinct cuproptosis patterns were generated. These two patterns were consistent with inflamed and noninflamed TME phenotypes and had potential role for predicting immunotherapy efficacies. We constructed a CRS for predicting individual patient's prognosis with high accuracy in TCGA-BLCA. Importantly, this CRS could be well validated in external cohorts including GSE32894 and GSE13507. Then, we developed a cuproptosis signature and found it was significantly negative correlated with tumor-infiltrating lymphocytes (TILs) both in TCGA-BLCA and Xiangya cohorts. Moreover, we revealed that patients in the high cuproptosis signature group represented a noninflamed TME phenotype on the single cell level. As expected, patients in the high cuproptosis signature group showed less sensitive to immunotherapy. Finally, we found that the high and low cuproptosis signature groups were consistent with luminal and basal subtypes of BLCA respectively, which validated the role of signature in TME in terms of molecular subtypes. CONCLUSIONS Cuproptosis patterns depict different TME phenotypes in BLCA. Our CRS and cuproptosis signature have potential role for predicting prognosis and immunotherapy efficacy, which might guide precise medicine.
Collapse
Affiliation(s)
- Huihuang Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xiongbing Zu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jiao Hu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zicheng Xiao
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhiyong Cai
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ning Gao
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- Department of Urology, Xiangya Boai Hospital, Changsha, China
| | - Jinbo Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
26
|
Laurentino TDS, Soares RDS, Marie SKN, Oba-Shinjo SM. Correlation of Matrisome-Associatted Gene Expressions with LOX Family Members in Astrocytomas Stratified by IDH Mutation Status. Int J Mol Sci 2022; 23:ijms23179507. [PMID: 36076905 PMCID: PMC9455728 DOI: 10.3390/ijms23179507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/02/2022] [Accepted: 08/16/2022] [Indexed: 12/05/2022] Open
Abstract
Tumor cell infiltrative ability into surrounding brain tissue is a characteristic of diffusely infiltrative astrocytoma and is strongly associated with extracellular matrix (ECM) stiffness. Collagens are the most abundant ECM scaffolding proteins and contribute to matrix organization and stiffness. LOX family members, copper-dependent amine oxidases, participate in the collagen and elastin crosslinking that determine ECM tensile strength. Common IDH mutations in lower-grade gliomas (LGG) impact prognosis and have been associated with ECM stiffness. We analyzed the expression levels of LOX family members and matrisome-associated genes in astrocytoma stratified by malignancy grade and IDH mutation status. A progressive increase in expression of all five LOX family members according to malignancy grade was found. LOX, LOXL1, and LOXL3 expression correlated with matrisome gene expressions. LOXL1 correlations were detected in LGG with IDH mutation (IDHmut), LOXL3 correlations in LGG with IDH wild type (IDHwt) and strong LOX correlations in glioblastoma (GBM) were found. These increasing correlations may explain the increment of ECM stiffness and tumor aggressiveness from LGG-IDHmut and LGG-IDHwt through to GBM. The expression of the mechanosensitive transcription factor, β-catenin, also increased with malignancy grade and was correlated with LOXL1 and LOXL3 expression, suggesting involvement of this factor in the outside–in signaling pathway.
Collapse
|
27
|
Wei J, Yao J, Yan M, Xie Y, Liu P, Mao Y, Li X. The role of matrix stiffness in cancer stromal cell fate and targeting therapeutic strategies. Acta Biomater 2022; 150:34-47. [DOI: 10.1016/j.actbio.2022.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/11/2022] [Accepted: 08/02/2022] [Indexed: 11/15/2022]
|
28
|
Mechanistic insight into lysyl oxidase in vascular remodeling and angiogenesis. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
29
|
Perryman L, Gray SG. Fibrosis in Mesothelioma: Potential Role of Lysyl Oxidases. Cancers (Basel) 2022; 14:981. [PMID: 35205728 PMCID: PMC8870010 DOI: 10.3390/cancers14040981] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 12/13/2022] Open
Abstract
Immunotherapies (such as checkpoint inhibitors) and standard chemotherapies (such as cisplatin) have limitations in the successful treatment of malignant pleural mesothelioma (MPM). Fibrosis is the accumulation of collagen in the extracellular matrix (ECM) of tissues, making them denser than that of healthy tissues and thereby affecting drug delivery and immune cell infiltration. Moreover, fibrosis severely affects the patient's breathing and quality of life. The production of collagen and its assembly is highly regulated by various enzymes such as lysyl oxidases. Many solid tumors aberrantly express the family of lysyl oxidases (LOX/LOXL). This review examines how LOX/LOXLs were found to be dysregulated in noncancerous and cancerous settings, discusses their roles in solid tumor fibrosis and pathogenesis and explores the role of fibrosis in the development and poor clinical outcomes of patients with MPM. We examine the current preclinical status of drugs targeting LOX/LOXLs and how the incorporation of such drugs may have therapeutic benefits in the treatment and management of patients with MPM.
Collapse
Affiliation(s)
- Lara Perryman
- Drug Discovery Department, Pharmaxis Ltd., Sydney, NSW 2086, Australia;
| | - Steven G. Gray
- Thoracic Oncology, Labmed Directorate, St James’s Hospital, D08 RX0X Dublin, Ireland
| |
Collapse
|
30
|
Chen X, Margaret C, Hicks MJ, Sarkar P, Gaber MW, Man TK. LOX upregulates FAK phosphorylation to promote metastasis in osteosarcoma. Genes Dis 2022; 10:254-266. [PMID: 37013056 PMCID: PMC10066266 DOI: 10.1016/j.gendis.2021.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 12/02/2021] [Accepted: 12/21/2021] [Indexed: 10/19/2022] Open
Abstract
Osteosarcoma is a malignant bone tumor that commonly occurs in the pediatric population. Despite the use of chemotherapy and surgery, metastasis remains to be the leading cause of death in patients with osteosarcoma. We have previously reported that cytoplasmic mislocalization of p27 is associated with a poor outcome in osteosarcoma. In this study, we further show that lysyl oxidase (LOX) expression was associated with p27 mislocalization. LOX is an enigmatic molecule that acts as a tumor suppressor or a metastatic promoter; however, its role in osteosarcoma is still unclear. Hence, we performed both in vitro and in vivo analyses to dissect the role of LOX in osteosarcoma. The result of our survival analysis indicated that LOX expression significantly correlated with a poor outcome in osteosarcoma with or without controlling for the initial metastasis status (P < 0.05). Functionally, we found that higher LOX expression promoted osteosarcoma cell proliferation, migration, and invasiveness in vitro and produced a higher number of mice with pulmonary metastases in an orthotopic xenograft mouse model. Mechanistically, phospho-FAK was increased in osteosarcoma cells with high LOX expression. Our results further showed that FAK inhibition significantly reduced tumor cell proliferation and migration in vitro as well as LOX-mediated metastasis in mice. Together, our findings suggest that there is a novel link between p27 mislocalization and LOX expression. LOX plays a pivotal role in osteosarcoma metastasis by upregulating FAK phosphorylation. FAK inhibition may constitute a promising therapeutic strategy to reduce the development of metastasis in osteosarcoma with LOX overexpression.
Collapse
|
31
|
Serum Lysyl Oxidase Levels and Lysyl Oxidase Gene Polymorphism in Ovarian Cancer Patients of Eastern Indian Population. Diagnostics (Basel) 2021; 12:diagnostics12010053. [PMID: 35054220 PMCID: PMC8774920 DOI: 10.3390/diagnostics12010053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/18/2021] [Accepted: 12/06/2021] [Indexed: 11/17/2022] Open
Abstract
(1) Background: Lysyl oxidase (LOX) plays a dual role in carcinogenesis and studies show a higher risk of cancer in LOX G473A variants. The present study evaluated the pattern of LOX G473A polymorphism (rs1800449) and serum LOX levels in ovarian cancer patients. (2) Methods: Serum LOX levels were estimated by enzyme linked immunosorbent assay (ELISA). A polymorphism of rs1800449 of LOX gene was detected by the polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method. Selected samples were sequenced for external validation. (3) Results: A majority of study participants were from low socio-economic status. Serum LOX level was significantly higher in ovarian cancer patients as compared to control. Serum LOX level in early-stage ovarian cancer was significantly lower as compared to advanced stage (FIGO stage III & IV). Wild type GG genotype was used as reference. Genotypes AA were associated with a significant risk of epithelial ovarian cancer (OR 3.208; p value- 0.033). A allele of rs1800449 polymorphism of LOX gene, the odds ratio was 1.866 (95% Confidence Interval 1.112–3.16) p value = 0.017 (4) Conclusions: A allele of rs1800449 polymorphism of LOX gene presents an increased risk of ovarian cancer in East Indian population. Serum LOX levels could be a potential biomarker for the diagnosis and prognosis of ovarian cancer.
Collapse
|
32
|
Karamanos NK, Theocharis AD, Piperigkou Z, Manou D, Passi A, Skandalis SS, Vynios DH, Orian-Rousseau V, Ricard-Blum S, Schmelzer CEH, Duca L, Durbeej M, Afratis NA, Troeberg L, Franchi M, Masola V, Onisto M. A guide to the composition and functions of the extracellular matrix. FEBS J 2021; 288:6850-6912. [PMID: 33605520 DOI: 10.1111/febs.15776] [Citation(s) in RCA: 482] [Impact Index Per Article: 120.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/13/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022]
Abstract
Extracellular matrix (ECM) is a dynamic 3-dimensional network of macromolecules that provides structural support for the cells and tissues. Accumulated knowledge clearly demonstrated over the last decade that ECM plays key regulatory roles since it orchestrates cell signaling, functions, properties and morphology. Extracellularly secreted as well as cell-bound factors are among the major members of the ECM family. Proteins/glycoproteins, such as collagens, elastin, laminins and tenascins, proteoglycans and glycosaminoglycans, hyaluronan, and their cell receptors such as CD44 and integrins, responsible for cell adhesion, comprise a well-organized functional network with significant roles in health and disease. On the other hand, enzymes such as matrix metalloproteinases and specific glycosidases including heparanase and hyaluronidases contribute to matrix remodeling and affect human health. Several cell processes and functions, among them cell proliferation and survival, migration, differentiation, autophagy, angiogenesis, and immunity regulation are affected by certain matrix components. Structural alterations have been also well associated with disease progression. This guide on the composition and functions of the ECM gives a broad overview of the matrisome, the major ECM macromolecules, and their interaction networks within the ECM and with the cell surface, summarizes their main structural features and their roles in tissue organization and cell functions, and emphasizes the importance of specific ECM constituents in disease development and progression as well as the advances in molecular targeting of ECM to design new therapeutic strategies.
Collapse
Affiliation(s)
- Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Achilleas D Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Dimitra Manou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Spyros S Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Demitrios H Vynios
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Véronique Orian-Rousseau
- Karlsruhe Institute of Technology, Institute of Biological and Chemical Systems- Functional Molecular Systems, Eggenstein-Leopoldshafen, Germany
| | - Sylvie Ricard-Blum
- University of Lyon, UMR 5246, ICBMS, Université Lyon 1, CNRS, Villeurbanne Cedex, France
| | - Christian E H Schmelzer
- Fraunhofer Institute for Microstructure of Materials and Systems IMWS, Halle (Saale), Germany
- Institute of Pharmacy, Faculty of Natural Sciences I, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Laurent Duca
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2: Matrix Aging and Vascular Remodelling, Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Madeleine Durbeej
- Department of Experimental Medical Science, Unit of Muscle Biology, Lund University, Sweden
| | - Nikolaos A Afratis
- Department Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Linda Troeberg
- Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, Norwich, UK
| | - Marco Franchi
- Department for Life Quality Study, University of Bologna, Rimini, Italy
| | | | - Maurizio Onisto
- Department of Biomedical Sciences, University of Padova, Italy
| |
Collapse
|
33
|
Yang A, Yan X, Xu H, Fan X, Zhang M, Huang T, Li W, Chen W, Jia J, You H. Selective depletion of hepatic stellate cells-specific LOXL1 alleviates liver fibrosis. FASEB J 2021; 35:e21918. [PMID: 34569648 DOI: 10.1096/fj.202100374r] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 08/06/2021] [Accepted: 08/27/2021] [Indexed: 12/13/2022]
Abstract
The role of LOXL1 in fibrosis via mediating ECM crosslinking and stabilization is well established; however, the role of hepatic stellate cells (HSCs)-specific LOXL1 in the development of fibrosis remains unknown. We generated HSCs-specific Loxl1-depleted mice (Loxl1Gfap-cre mice) to investigate the HSCs-specific contribution of LOXL1 in the pathogenesis of fibrosis. Loxl1fl/fl mice were used as the control. Furthermore, we used RNA sequencing to explore the underlying changes in the transcriptome. Results of the sirius red staining, type I collagen immunolabeling, and hydroxyproline content analysis, coupled with the reduced expression of profibrogenic genes revealed that Loxl1Gfap-cre mice with CCl4 -induced fibrosis exhibited decreased hepatic fibrosis. In addition, Loxl1Gfap-cre mice exhibited reduced macrophage tissue infiltration by CD68-positive cells and decreased expression of inflammatory genes compared with the controls. RNA sequencing identified integrin α8 (ITGA8) as a key modulator of LOXL1-mediated liver fibrosis. Functional analyses showed that siRNA silencing of Itga8 in cultured fibroblasts led to a decline in the LOXL1 expression and inhibition of fibroblast activation. Mechanistic analyses indicated that LOXL1 activated the FAK/PI3K/AKT/HIF1a signaling pathway, and the addition of inhibitors of FAK or PI3K reversed these results via downregulation of LOXL1. Furthermore, HIF1a directly interacted with LOXL1 and upregulated its expression, indicating that LOXL1 can positively self-regulate by forming a positive feedback loop with the FAK/PI3K/AKT/HIF1a pathway. We demonstrated that HSCs-specific Loxl1 deficiency prevented fibrosis, inflammation and that ITGA8/FAK/PI3K/AKT/HIF1a was essential for the function and expression of LOXL1. Knowledge of this approach can provide novel mechanisms and targets to treat fibrosis in the future.
Collapse
Affiliation(s)
- Aiting Yang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China.,Beijing Clinical Medicine Institute, Beijing, P.R. China.,National Clinical Research Center of Digestive Diseases, Beijing, P.R. China
| | - Xuzhen Yan
- National Clinical Research Center of Digestive Diseases, Beijing, P.R. China.,Liver Research Center, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| | - Hufeng Xu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China.,Beijing Clinical Medicine Institute, Beijing, P.R. China.,National Clinical Research Center of Digestive Diseases, Beijing, P.R. China
| | - Xu Fan
- National Clinical Research Center of Digestive Diseases, Beijing, P.R. China.,Liver Research Center, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| | - Mengyang Zhang
- National Clinical Research Center of Digestive Diseases, Beijing, P.R. China.,Liver Research Center, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| | - Tao Huang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China.,Beijing Clinical Medicine Institute, Beijing, P.R. China.,National Clinical Research Center of Digestive Diseases, Beijing, P.R. China
| | - Weiyu Li
- National Clinical Research Center of Digestive Diseases, Beijing, P.R. China.,Liver Research Center, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| | - Wei Chen
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China.,Beijing Clinical Medicine Institute, Beijing, P.R. China.,National Clinical Research Center of Digestive Diseases, Beijing, P.R. China
| | - Jidong Jia
- Beijing Clinical Medicine Institute, Beijing, P.R. China.,National Clinical Research Center of Digestive Diseases, Beijing, P.R. China.,Liver Research Center, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| | - Hong You
- Beijing Clinical Medicine Institute, Beijing, P.R. China.,National Clinical Research Center of Digestive Diseases, Beijing, P.R. China.,Liver Research Center, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| |
Collapse
|
34
|
Martinez-Vidal L, Murdica V, Venegoni C, Pederzoli F, Bandini M, Necchi A, Salonia A, Alfano M. Causal contributors to tissue stiffness and clinical relevance in urology. Commun Biol 2021; 4:1011. [PMID: 34446834 PMCID: PMC8390675 DOI: 10.1038/s42003-021-02539-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 08/02/2021] [Indexed: 02/07/2023] Open
Abstract
Mechanomedicine is an emerging field focused on characterizing mechanical changes in cells and tissues coupled with a specific disease. Understanding the mechanical cues that drive disease progression, and whether tissue stiffening can precede disease development, is crucial in order to define new mechanical biomarkers to improve and develop diagnostic and prognostic tools. Classically known stromal regulators, such as fibroblasts, and more recently acknowledged factors such as the microbiome and extracellular vesicles, play a crucial role in modifications to the stroma and extracellular matrix (ECM). These modifications ultimately lead to an alteration of the mechanical properties (stiffness) of the tissue, contributing to disease onset and progression. We describe here classic and emerging mediators of ECM remodeling, and discuss state-of-the-art studies characterizing mechanical fingerprints of urological diseases, showing a general trend between increased tissue stiffness and severity of disease. Finally, we point to the clinical potential of tissue stiffness as a diagnostic and prognostic factor in the urological field, as well as a possible target for new innovative drugs.
Collapse
Affiliation(s)
- Laura Martinez-Vidal
- Vita-Salute San Raffaele University, Milan, Italy.
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS San Raffaele Hospital, Milan, Italy.
| | - Valentina Murdica
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS San Raffaele Hospital, Milan, Italy
| | - Chiara Venegoni
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS San Raffaele Hospital, Milan, Italy
| | - Filippo Pederzoli
- Vita-Salute San Raffaele University, Milan, Italy
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS San Raffaele Hospital, Milan, Italy
| | - Marco Bandini
- Vita-Salute San Raffaele University, Milan, Italy
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS San Raffaele Hospital, Milan, Italy
| | | | - Andrea Salonia
- Vita-Salute San Raffaele University, Milan, Italy
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS San Raffaele Hospital, Milan, Italy
| | - Massimo Alfano
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS San Raffaele Hospital, Milan, Italy
| |
Collapse
|
35
|
Marzo T, La Mendola D. The Effects on Angiogenesis of Relevant Inorganic Chemotherapeutics. Curr Top Med Chem 2021; 21:73-86. [PMID: 33243124 DOI: 10.2174/1568026620666201126163436] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022]
Abstract
Angiogenesis is a key process allowing the formation of blood vessels. It is crucial for all the tissues and organs, ensuring their function and growth. Angiogenesis is finely controlled by several mechanisms involving complex interactions between pro- or antiangiogenic factors, and an imbalance in this control chain may result in pathological conditions. Metals as copper, zinc and iron cover an essential role in regulating angiogenesis, thus therapies having physiological metals as target have been proposed. In addition, some complexes of heavier metal ions (e.g., Pt, Au, Ru) are currently used as established or experimental anticancer agents targeting genomic or non-genomic targets. These molecules may affect the angiogenic mechanisms determining different effects that have been only poorly and non-systematically investigated so far. Accordingly, in this review article, we aim to recapitulate the impact on the angiogenic process of some reference anticancer drugs, and how it is connected to the overall pharmacological effects. In addition, we highlight how the activity of these drugs can be related to the role of biological essential metal ions. Overall, this may allow a deeper description and understanding of the antineoplastic activity of both approved or experimental metal complexes, providing important insights for the synthesis of new inorganic drugs able to overcome resistance and recurrence phenomena.
Collapse
Affiliation(s)
- Tiziano Marzo
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| | - Diego La Mendola
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| |
Collapse
|
36
|
Micalet A, Moeendarbary E, Cheema U. 3D In Vitro Models for Investigating the Role of Stiffness in Cancer Invasion. ACS Biomater Sci Eng 2021. [PMID: 34081437 DOI: 10.1021/acsbiomaterials.0c01530] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Tumorigenesis is attributed to the interactions of cancer cells with the tumor microenvironment through both biochemical cues and physical stimuli. Increased matrix deposition and realignment of the collagen fibers are detected by cancer cells, inducing epithelial-to-mesenchymal transition, which in turn stimulates cell motility and invasiveness. METHODS This review provides an overview of current research on the role of the physical microenvironment in cancer invasion. This was achieved by using a systematic approach and providing meta-analyses. Particular focus was placed on in vitro three-dimensional models of epithelial cancers. We investigated questions such as the effect of matrix stiffening, activation of stromal cells, and identified potential advances in mechano-based therapies. RESULTS Meta-analysis revealed that 64% of studies report cancer invasion promotion as stiffness increases, while 36% report the opposite. Experimental approaches and data interpretations were varied, each affecting the invasion of cancer differently. Examples are the experimental timeframes used (24 h to 21 days), the type of polymer used (24 types), and choice of cell line (33 cell lines). The stiffness of the 3D matrices varied from 0.5 to 300 kPa and 19% of these matrices' stiffness were outside commonly accepted physiological range. 100% of the studies outside biological stiffness range (above 20 kPa) report that stiffness does not promote cancer invasion. CONCLUSIONS Taking this analysis into account, we inform on the type of experimental approaches that could be the most relevant and provide what would be a standardized protocol and reporting strategy.
Collapse
Affiliation(s)
- Auxtine Micalet
- Department of Mechanical Engineering, University College London (UCL), Torrington Place, London, U.K. WC1E 6BT.,Division of Surgery and Interventional Sciences, UCL Centre for 3D Models of Health and Disease, University College London (UCL), Charles Bell House, London, U.K. W1W 7TS
| | - Emad Moeendarbary
- Department of Mechanical Engineering, University College London (UCL), Torrington Place, London, U.K. WC1E 6BT.,Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139, United States
| | - Umber Cheema
- Division of Surgery and Interventional Sciences, UCL Centre for 3D Models of Health and Disease, University College London (UCL), Charles Bell House, London, U.K. W1W 7TS
| |
Collapse
|
37
|
Collagen molecular phenotypic switch between non-neoplastic and neoplastic canine mammary tissues. Sci Rep 2021; 11:8659. [PMID: 33883562 PMCID: PMC8060395 DOI: 10.1038/s41598-021-87380-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/23/2021] [Indexed: 01/24/2023] Open
Abstract
In spite of major advances over the past several decades in diagnosis and treatment, breast cancer remains a global cause of morbidity and premature death for both human and veterinary patients. Due to multiple shared clinicopathological features, dogs provide an excellent model of human breast cancer, thus, a comparative oncology approach may advance our understanding of breast cancer biology and improve patient outcomes. Despite an increasing awareness of the critical role of fibrillar collagens in breast cancer biology, tumor-permissive collagen features are still ill-defined. Here, we characterize the molecular and morphological phenotypes of type I collagen in canine mammary gland tumors. Canine mammary carcinoma samples contained longer collagen fibers as well as a greater population of wider fibers compared to non-neoplastic and adenoma samples. Furthermore, the total number of collagen cross-links enriched in the stable hydroxylysine-aldehyde derived cross-links was significantly increased in neoplastic mammary gland samples compared to non-neoplastic mammary gland tissue. The mass spectrometric analyses of type I collagen revealed that in malignant mammary tumor samples, lysine residues, in particular those in the telopeptides, were markedly over-hydroxylated in comparison to non-neoplastic mammary tissue. The extent of glycosylation of hydroxylysine residues was comparable among the groups. Consistent with these data, expression levels of genes encoding lysyl hydroxylase 2 (LH2) and its molecular chaperone FK506-binding protein 65 were both significantly increased in neoplastic samples. These alterations likely lead to an increase in the LH2-mediated stable collagen cross-links in mammary carcinoma that may promote tumor cell metastasis in these patients.
Collapse
|
38
|
Targeting Lysyl Oxidase Family Meditated Matrix Cross-Linking as an Anti-Stromal Therapy in Solid Tumours. Cancers (Basel) 2021; 13:cancers13030491. [PMID: 33513979 PMCID: PMC7865543 DOI: 10.3390/cancers13030491] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary To improve efficacy of solid cancer treatment, efforts have shifted towards targeting both the cancer cells and the surrounding tumour tissue they grow in. The lysyl oxidase (LOX) family of enzymes underpin the fibrotic remodeling of the tumour microenvironment to promote both cancer growth, spread throughout the body and modulate response to therapies. This review examines how the lysyl oxidase family is involved in tumour development, how they can be targeted, and their potential as diagnostic and prognostic biomarkers in solid tumours. Abstract The lysyl oxidase (LOX) family of enzymes are a major driver in the biogenesis of desmoplastic matrix at the primary tumour and secondary metastatic sites. With the increasing interest in and development of anti-stromal therapies aimed at improving clinical outcomes of cancer patients, the Lox family has emerged as a potentially powerful clinical target. This review examines how lysyl oxidase family dysregulation in solid cancers contributes to disease progression and poor patient outcomes, as well as an evaluation of the preclinical landscape of LOX family targeting therapeutics. We also discuss the suitability of the LOX family as a diagnostic and/or prognostic marker in solid tumours.
Collapse
|
39
|
Yang H, Kuo YH, Smith ZI, Spangler J. Targeting cancer metastasis with antibody therapeutics. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1698. [PMID: 33463090 DOI: 10.1002/wnan.1698] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 12/12/2022]
Abstract
Cancer metastasis, the spread of disease from a primary to a distal site through the circulatory or lymphatic systems, accounts for over 90% of all cancer related deaths. Despite significant progress in the field of cancer therapy in recent years, mortality rates remain dramatically higher for patients with metastatic disease versus those with local or regional disease. Although there is clearly an urgent need to develop drugs that inhibit cancer spread, the overwhelming majority of anticancer therapies that have been developed to date are designed to inhibit tumor growth but fail to address the key stages of the metastatic process: invasion, intravasation, circulation, extravasation, and colonization. There is growing interest in engineering targeted therapeutics, such as antibody drugs, that inhibit various steps in the metastatic cascade. We present an overview of antibody therapeutic approaches, both in the pipeline and in the clinic, that disrupt the essential mechanisms that underlie cancer metastasis. These therapies include classes of antibodies that indirectly target metastasis, including anti-integrin, anticadherin, and immune checkpoint blocking antibodies, as well as monoclonal and bispecific antibodies that are specifically designed to interrupt disease dissemination. Although few antimetastatic antibodies have achieved clinical success to date, there are many promising candidates in various stages of development, and novel targets and approaches are constantly emerging. Collectively, these efforts will enrich our understanding of the molecular drivers of metastasis, and the new strategies that arise promise to have a profound impact on the future of cancer therapeutic development. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Huilin Yang
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Yun-Huai Kuo
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Zion I Smith
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jamie Spangler
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
40
|
Vallet SD, Berthollier C, Salza R, Muller L, Ricard-Blum S. The Interactome of Cancer-Related Lysyl Oxidase and Lysyl Oxidase-Like Proteins. Cancers (Basel) 2020; 13:E71. [PMID: 33383846 PMCID: PMC7794802 DOI: 10.3390/cancers13010071] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 12/22/2020] [Indexed: 12/16/2022] Open
Abstract
The members of the lysyl oxidase (LOX) family are amine oxidases, which initiate the covalent cross-linking of the extracellular matrix (ECM), regulate ECM stiffness, and contribute to cancer progression. The aim of this study was to build the first draft of the interactome of the five members of the LOX family in order to determine its molecular functions, the biological and signaling pathways mediating these functions, the biological processes it is involved in, and if and how it is rewired in cancer. In vitro binding assays, based on surface plasmon resonance and bio-layer interferometry, combined with queries of interaction databases and interaction datasets, were used to retrieve interaction data. The interactome was then analyzed using computational tools. We identified 31 new interactions and 14 new partners of LOXL2, including the α5β1 integrin, and built an interactome comprising 320 proteins, 5 glycosaminoglycans, and 399 interactions. This network participates in ECM organization, degradation and cross-linking, cell-ECM interactions mediated by non-integrin and integrin receptors, protein folding and chaperone activity, organ and blood vessel development, cellular response to stress, and signal transduction. We showed that this network is rewired in colorectal carcinoma, leading to a switch from ECM organization to protein folding and chaperone activity.
Collapse
Affiliation(s)
- Sylvain D. Vallet
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, INSA Lyon, CPE, Institute of Molecular and Supramolecular Chemistry and Biochemistry, UMR 5246, F-69622 Villeurbanne CEDEX, France; (S.D.V.); (C.B.); (R.S.)
| | - Coline Berthollier
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, INSA Lyon, CPE, Institute of Molecular and Supramolecular Chemistry and Biochemistry, UMR 5246, F-69622 Villeurbanne CEDEX, France; (S.D.V.); (C.B.); (R.S.)
| | - Romain Salza
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, INSA Lyon, CPE, Institute of Molecular and Supramolecular Chemistry and Biochemistry, UMR 5246, F-69622 Villeurbanne CEDEX, France; (S.D.V.); (C.B.); (R.S.)
| | - Laurent Muller
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, 75231 Paris CEDEX 05, France;
| | - Sylvie Ricard-Blum
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, INSA Lyon, CPE, Institute of Molecular and Supramolecular Chemistry and Biochemistry, UMR 5246, F-69622 Villeurbanne CEDEX, France; (S.D.V.); (C.B.); (R.S.)
| |
Collapse
|
41
|
Niland S, Eble JA. Hold on or Cut? Integrin- and MMP-Mediated Cell-Matrix Interactions in the Tumor Microenvironment. Int J Mol Sci 2020; 22:ijms22010238. [PMID: 33379400 PMCID: PMC7794804 DOI: 10.3390/ijms22010238] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023] Open
Abstract
The tumor microenvironment (TME) has become the focus of interest in cancer research and treatment. It includes the extracellular matrix (ECM) and ECM-modifying enzymes that are secreted by cancer and neighboring cells. The ECM serves both to anchor the tumor cells embedded in it and as a means of communication between the various cellular and non-cellular components of the TME. The cells of the TME modify their surrounding cancer-characteristic ECM. This in turn provides feedback to them via cellular receptors, thereby regulating, together with cytokines and exosomes, differentiation processes as well as tumor progression and spread. Matrix remodeling is accomplished by altering the repertoire of ECM components and by biophysical changes in stiffness and tension caused by ECM-crosslinking and ECM-degrading enzymes, in particular matrix metalloproteinases (MMPs). These can degrade ECM barriers or, by partial proteolysis, release soluble ECM fragments called matrikines, which influence cells inside and outside the TME. This review examines the changes in the ECM of the TME and the interaction between cells and the ECM, with a particular focus on MMPs.
Collapse
|
42
|
Targeting Mechanotransduction in Osteosarcoma: A Comparative Oncology Perspective. Int J Mol Sci 2020; 21:ijms21207595. [PMID: 33066583 PMCID: PMC7589883 DOI: 10.3390/ijms21207595] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 12/13/2022] Open
Abstract
Mechanotransduction is the process in which cells can convert extracellular mechanical stimuli into biochemical changes within a cell. While this a normal process for physiological development and function in many organ systems, tumour cells can exploit this process to promote tumour progression. Here we summarise the current state of knowledge of mechanotransduction in osteosarcoma (OSA), the most common primary bone tumour, referencing both human and canine models and other similar mesenchymal malignancies (e.g., Ewing sarcoma). Specifically, we discuss the mechanical properties of OSA cells, the pathways that these cells utilise to respond to external mechanical cues, and mechanotransduction-targeting strategies tested in OSA so far. We point out gaps in the literature and propose avenues to address them. Understanding how the physical microenvironment influences cell signalling and behaviour will lead to the improved design of strategies to target the mechanical vulnerabilities of OSA cells.
Collapse
|
43
|
Gordon B, Gadi VK. The Role of the Tumor Microenvironment in Developing Successful Therapeutic and Secondary Prophylactic Breast Cancer Vaccines. Vaccines (Basel) 2020; 8:vaccines8030529. [PMID: 32937885 PMCID: PMC7565925 DOI: 10.3390/vaccines8030529] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/09/2020] [Accepted: 09/13/2020] [Indexed: 12/12/2022] Open
Abstract
Breast cancer affects roughly one in eight women over their lifetime and is a leading cause of cancer-related death in women. While outcomes have improved in recent years, prognosis remains poor for patients who present with either disseminated disease or aggressive molecular subtypes. Cancer immunotherapy has revolutionized the treatment of several cancers, with therapeutic vaccines aiming to direct the cytotoxic immune program against tumor cells showing particular promise. However, these results have yet to translate to breast cancer, which remains largely refractory from such approaches. Recent evidence suggests that the breast tumor microenvironment (TME) is an important and long understudied barrier to the efficacy of therapeutic vaccines. Through an improved understanding of the complex and biologically diverse breast TME, it may be possible to advance new combination strategies to render breast carcinomas sensitive to the effects of therapeutic vaccines. Here, we discuss past and present efforts to advance therapeutic vaccines in the treatment of breast cancer, the molecular mechanisms through which the TME contributes to the failure of such approaches, as well as the potential means through which these can be overcome.
Collapse
Affiliation(s)
- Benjamin Gordon
- Department of Physiology and Biophysics, University of Illinois College of Medicine, Chicago, IL 60612, USA
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, IL 60612, USA
- Correspondence:
| | - Vijayakrishna K. Gadi
- Division of Hematology and Oncology, University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA;
| |
Collapse
|
44
|
Yamauchi M, Gibbons DL, Zong C, Fradette JJ, Bota-Rabassedas N, Kurie JM. Fibroblast heterogeneity and its impact on extracellular matrix and immune landscape remodeling in cancer. Matrix Biol 2020; 91-92:8-18. [PMID: 32442601 DOI: 10.1016/j.matbio.2020.05.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 05/06/2020] [Accepted: 05/06/2020] [Indexed: 12/12/2022]
Abstract
Tumor progression is marked by dense collagenous matrix accumulations that dynamically reorganize to accommodate a growing and invasive tumor mass. Cancer-associated fibroblasts (CAFs) play an essential role in matrix remodeling and influence other processes in the tumor microenvironment, including angiogenesis, immunosuppression, and invasion. These findings have spawned efforts to elucidate CAF functionality at the single-cell level. Here, we will discuss how those efforts have impacted our understanding of the ways in which CAFs govern matrix remodeling and the influence of matrix remodeling on the development of an immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Mitsuo Yamauchi
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NS, United States
| | - Don L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, United States
| | - Chenghang Zong
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Jared J Fradette
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, United States
| | - Neus Bota-Rabassedas
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, United States
| | - Jonathan M Kurie
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, United States.
| |
Collapse
|
45
|
Zhuyan J, Chen M, Zhu T, Bao X, Zhen T, Xing K, Wang Q, Zhu S. Critical steps to tumor metastasis: alterations of tumor microenvironment and extracellular matrix in the formation of pre-metastatic and metastatic niche. Cell Biosci 2020; 10:89. [PMID: 32742634 PMCID: PMC7388444 DOI: 10.1186/s13578-020-00453-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 07/21/2020] [Indexed: 02/07/2023] Open
Abstract
For decades, cancer metastasis has been a heated topic for its high mortality. Previous research has shown that pre-metastatic niche and metastatic niche are the 2 crucial steps in cancer metastasis, assisting cancerous cells' infiltration, survival, and colonization at target sites. More recent studies have unraveled details about the specific mechanisms related to the modification of pro-invasion environments. Here, we will review literatures on extracellular matrix (ECM) alterations, general cancer metastasis, organ specificity, pre-metastatic niche, metastatic niche, colony formation and impact on the course of metastasis. Respectively, the metastatic mechanisms like effect of hypoxia or inflammation on pre-metastatic niche construction, as well as the interaction between cancer cells and local milieu will be discussed. Based on the evidences of metastatic niches, we revisit and discussed the "Seed and Soil" hypothesis by Paget. This review will seek to provide insight into the mechanism of metastatic organ specificity which pre-metastatic niche and metastatic niche might suggest from an evolutionary aspect.
Collapse
Affiliation(s)
- Jianan Zhuyan
- School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438 China
- Shanghai Starriver Bilingual School, Shanghai, 201100 China
| | - Mingyu Chen
- Department of Neurosurgery, Huashan Hospital, Shanghai, 200040 China
| | - Tianhao Zhu
- Shanghai Starriver Bilingual School, Shanghai, 201100 China
| | - Xunxia Bao
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093 China
| | - Timing Zhen
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093 China
| | - Kaichen Xing
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093 China
| | - Qiubo Wang
- Department of Clinical Laboratory, Wuxi 9th Affiliated Hospital of Soochow University, No.999 Liangxi Road, Wuxi, China
| | - Sibo Zhu
- School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai, 200438 China
| |
Collapse
|
46
|
D'Angelo E, Lindoso RS, Sensi F, Pucciarelli S, Bussolati B, Agostini M, Collino F. Intrinsic and Extrinsic Modulators of the Epithelial to Mesenchymal Transition: Driving the Fate of Tumor Microenvironment. Front Oncol 2020; 10:1122. [PMID: 32793478 PMCID: PMC7393251 DOI: 10.3389/fonc.2020.01122] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/04/2020] [Indexed: 12/11/2022] Open
Abstract
The epithelial to mesenchymal transition (EMT) is an evolutionarily conserved process. In cancer, EMT can activate biochemical changes in tumor cells that enable the destruction of the cellular polarity, leading to the acquisition of invasive capabilities. EMT regulation can be triggered by intrinsic and extrinsic signaling, allowing the tumor to adapt to the microenvironment demand in the different stages of tumor progression. In concomitance, tumor cells undergoing EMT actively interact with the surrounding tumor microenvironment (TME) constituted by cell components and extracellular matrix as well as cell secretome elements. As a result, the TME is in turn modulated by the EMT process toward an aggressive behavior. The current review presents the intrinsic and extrinsic modulators of EMT and their relationship with the TME, focusing on the non-cell-derived components, such as secreted metabolites, extracellular matrix, as well as extracellular vesicles. Moreover, we explore how these modulators can be suitable targets for anticancer therapy and personalized medicine.
Collapse
Affiliation(s)
- Edoardo D'Angelo
- First Surgical Clinic, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
- LIFELAB Program, Consorzio per la Ricerca Sanitaria–CORIS, Veneto Region, Padua, Italy
- Institute of Pediatric Research, Fondazione Citta della Speranza, Padua, Italy
| | - Rafael Soares Lindoso
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine–REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Francesca Sensi
- Institute of Pediatric Research, Fondazione Citta della Speranza, Padua, Italy
- Department of Molecular Sciences and Nanosystems, Cà Foscari University of Venice, Venice, Italy
| | - Salvatore Pucciarelli
- First Surgical Clinic, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Benedetta Bussolati
- Department of Medical Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Marco Agostini
- First Surgical Clinic, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
- LIFELAB Program, Consorzio per la Ricerca Sanitaria–CORIS, Veneto Region, Padua, Italy
- Institute of Pediatric Research, Fondazione Citta della Speranza, Padua, Italy
| | - Federica Collino
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Biomedical Sciences, University of Padova, Padua, Italy
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione Ca' Granda, IRCCS Policlinico di Milano, Milan, Italy
| |
Collapse
|
47
|
Laczko R, Csiszar K. Lysyl Oxidase (LOX): Functional Contributions to Signaling Pathways. Biomolecules 2020; 10:biom10081093. [PMID: 32708046 PMCID: PMC7465975 DOI: 10.3390/biom10081093] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/12/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Cu-dependent lysyl oxidase (LOX) plays a catalytic activity-related, primary role in the assembly of the extracellular matrix (ECM), a dynamic structural and regulatory framework which is essential for cell fate, differentiation and communication during development, tissue maintenance and repair. LOX, additionally, plays both activity-dependent and independent extracellular, intracellular and nuclear roles that fulfill significant functions in normal tissues, and contribute to vascular, cardiac, pulmonary, dermal, placenta, diaphragm, kidney and pelvic floor disorders. LOX activities have also been recognized in glioblastoma, diabetic neovascularization, osteogenic differentiation, bone matrix formation, ligament remodeling, polycystic ovary syndrome, fetal membrane rupture and tumor progression and metastasis. In an inflammatory context, LOX plays a role in diminishing pluripotent mesenchymal cell pools which are relevant to the pathology of diabetes, osteoporosis and rheumatoid arthritis. Most of these conditions involve mechanisms with complex cell and tissue type-specific interactions of LOX with signaling pathways, not only as a regulatory target, but also as an active player, including LOX-mediated alterations of cell surface receptor functions and mutual regulatory activities within signaling loops. In this review, we aim to provide insight into the diverse ways in which LOX participates in signaling events, and explore the mechanistic details and functional significance of the regulatory and cross-regulatory interactions of LOX with the EGFR, PDGF, VEGF, TGF-β, mechano-transduction, inflammatory and steroid signaling pathways.
Collapse
|
48
|
Wei S, Gao L, Wu C, Qin F, Yuan J. Role of the lysyl oxidase family in organ development (Review). Exp Ther Med 2020; 20:163-172. [PMID: 32536990 PMCID: PMC7282176 DOI: 10.3892/etm.2020.8731] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 03/02/2020] [Indexed: 02/05/2023] Open
Abstract
Lysyl oxidase proteins (LOXs) are amine oxidases, which are mainly located in smooth muscle cells and fibroblasts and serve an important role in the formation of the extracellular matrix (ECM) in a copper-dependent manner. Owing to the ability of LOX proteins to modulate crosslinking between collagens and to promote the deposition of other fibers, they serve crucially in organogenesis and the subsequent organ development, as well as disease initiation and progression. In addition, ECM formation significantly influences organ morphological formation in both cancer- and non-tumor-related diseases, in addition to cellular epigenetic transformation and migration, under the influence of LOXs. A number of different signaling pathways regulate the LOXs expression and their enzymatic activation. The tissue remodeling and transformation process shares some resemblance between oncogenesis and embryogenesis. Additionally the roles that LOXs serve appeared to be stressed during oncogenesis and tumor metastasis. It has also been indicated LOXs have a noteworthy role in non-tumor diseases. Nonetheless, the role of LOXs in systemic or local organ development and disease control remains unknown. In the present study, the essential roles that LOXs play in embryogenesis were unveiled partially, whereas the role of LOXs in organ or systematic development requires further investigations. The present review aimed to discuss the roles of members of the LOX family in the context of the remodeling of organogenesis and organ development. In addition, the consequences of the malfunction of these proteins related to the development of abnormalities and resulting diseases is discussed.
Collapse
Affiliation(s)
- Shanzun Wei
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China.,Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Liang Gao
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China.,Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Changjing Wu
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Feng Qin
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jiuhong Yuan
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China.,Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
49
|
Rafaeva M, Erler JT. Framing cancer progression: influence of the organ- and tumour-specific matrisome. FEBS J 2020; 287:1454-1477. [PMID: 31972068 DOI: 10.1111/febs.15223] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/16/2019] [Accepted: 01/20/2020] [Indexed: 12/19/2022]
Abstract
The extracellular matrix (ECM) plays a crucial role in regulating organ homeostasis. It provides mechanical and biochemical cues directing cellular behaviour and, therefore, has control over the progression of diseases such as cancer. Recent efforts have greatly enhanced our knowledge of the protein composition of the ECM and its regulators, the so-called matrisome, in healthy and cancerous tissues; yet, an overview of the common signatures and organ-specific ECM in cancer is missing. Here, we address this by taking a detailed approach to review why cancer grows in certain organs, and focus on the influence of the matrisome at primary and metastatic tumour sites. Our in-depth and comprehensive review of the current literature and general understanding identifies important commonalities and distinctions, providing insight into the biology of metastasis, which could pave the way to improve future diagnostics and therapies.
Collapse
Affiliation(s)
- Maria Rafaeva
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen (UCPH), Denmark
| | - Janine T Erler
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen (UCPH), Denmark
| |
Collapse
|
50
|
Cai Q, Wang X, Wang S, Jin L, Ding J, Zhou D, Ma F. Gallbladder Cancer Progression Is Reversed by Nanomaterial-Induced Photothermal Therapy in Combination with Chemotherapy and Autophagy Inhibition. Int J Nanomedicine 2020; 15:253-262. [PMID: 32021178 PMCID: PMC6970248 DOI: 10.2147/ijn.s231289] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 01/02/2020] [Indexed: 12/20/2022] Open
Abstract
Introduction Gallbladder cancer (GBC) is the most common malignancy in biliary tract with extremely poor prognosis. Photothermal therapy (PTT) shows great promises for tumor therapy, which causes tumor cell death via selectively directed heating released by nanoparticles under the near-infrared irradiation. Through degrading damaged organelles and misfolded proteins in autophagosomes, autophagy plays a vital role in maintaining the intracellular homeostasis. The present study attempted to combine chemotherapy and autophagy blocking with PTT. Materials and Methods We purchased multi-walled carbon nanotubes from Nanostructured and Amorphous Materials and performed PTT using an 808-nm diode laser. The cytotoxic effects of PTT and chemotherapy in vitro were assessed by cell viability analysis. The effects of PTT and chemotherapy on autophagy in vitro were assessed by GFP-LC3 and Western blot. And these results were confirmed by in vivo experiment. Results Both PTT and chemotherapy could trigger cytoprotective autophagy to tolerate the cellular stresses and prolong the survival of GBC cell; therefore, the blocking of autophagy could enhance the efficacy of PTT and chemotherapy in GBC treatment in vitro and in vivo. Conclusion Chemotherapeutic drug doxorubicin and autophagy inhibitor chloroquine could enhance the efficacy of nanoparticle-mediated hyperthermia in GBC.
Collapse
Affiliation(s)
- Qiang Cai
- Department of General Surgery, Xinhua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200092, People's Republic of China.,Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Xinjing Wang
- Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Shouhua Wang
- Department of General Surgery, Xinhua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200092, People's Republic of China
| | - Longyang Jin
- Department of General Surgery, Xinhua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200092, People's Republic of China
| | - Jun Ding
- Department of Biliary and Pancreatic Surgery, Shanghai Shuguang Hospital Affiliated with Shanghai University of T.C.M., Shanghai 201203, People's Republic of China
| | - Di Zhou
- Department of General Surgery, Xinhua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200092, People's Republic of China
| | - Fei Ma
- Department of Oncology, Xinhua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai Institute for Pediatric Research, Shanghai 200092, People's Republic of China
| |
Collapse
|