1
|
Morataya-Reyes M, Villacorta A, Gutiérrez-García J, Egea R, Martín-Pérez J, Barguilla I, Marcos R, Hernández A. The long-term in vitro co-exposure of polyethylene terephthalate (PET) nanoplastics and cigarette smoke condensate exacerbates the induction of carcinogenic traits. JOURNAL OF HAZARDOUS MATERIALS 2025; 493:138359. [PMID: 40267709 DOI: 10.1016/j.jhazmat.2025.138359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/03/2025] [Accepted: 04/19/2025] [Indexed: 04/25/2025]
Abstract
This study examines the long-term impact of polyethylene terephthalate nanoplastics (PET-NPLs) and cigarette smoke condensate (CSC) on human lung BEAS-2B cells, focusing on key biological hallmarks of carcinogenesis. True-to-life PET-NPLs were generated from plastic water bottles and characterized to simulate environmental exposure conditions; and a comprehensive battery of assays was employed to assess genotoxicity, cellular transformation, and invasiveness. It was observed that, compared to passage control and individual exposures, co-exposure to PET-NPLs and CSC exacerbates oxidative stress, genotoxicity, and tumorigenic transformation, as evidenced by increased DNA damage, colony formation in soft agar, and enhanced cell migration and invasion. Transcriptomic analysis revealed a shift in cellular stress regulation including the upregulation of stress-response genes, including SLC7A11, NQO1, and HSPA1A, which are linked to oxidative stress adaptation and tumor survival. At the same time, key tumor-suppressor genes, such as LOX, and FN1, were significantly downregulated, promoting cellular transformation and invasiveness. These results provide compelling evidence that the combination of PET-NPLs and CSC enhances carcinogenic traits through oxidative stress, genomic instability, and disruption of tumor-suppressive pathways. This study underscores the importance of evaluating the synergistic effects of combined environmental exposures and their implications for human health.
Collapse
Affiliation(s)
- Michelle Morataya-Reyes
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Aliro Villacorta
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain; Facultad de Recursos Naturales Renovables, Universidad Arturo Prat, Iquique, Chile
| | - Javier Gutiérrez-García
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Raquel Egea
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Joan Martín-Pérez
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Irene Barguilla
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Ricard Marcos
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain.
| | - Alba Hernández
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain.
| |
Collapse
|
2
|
Çağlayan AD, Kahraman S, Çanakçı D, Tahtacı M, Altınboğa AA, Doğan HT. Association of lysyl oxidase expression with clinicopathological features in colorectal adenocarcinomas. Int J Colorectal Dis 2025; 40:75. [PMID: 40126681 PMCID: PMC11933219 DOI: 10.1007/s00384-025-04852-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/05/2025] [Indexed: 03/26/2025]
Abstract
PURPOSE Colorectal adenocarcinoma (CRC) is one of the leading causes of cancer-related mortality worldwide. Within the tumor microenvironment, neoplastic cells, along with tumor-promoting fibroblasts, contribute to the progression of CRC. Lysyl oxidase (LOX), an enzyme involved in this process facilitates collagen cross-linking within the extracellular matrix and plays a crucial role in remodeling the tumor microenvironment (TME) and promoting metastasis through epithelial-mesenchymal transition (EMT). This study investigates LOX expression in both tumor cells and the tumor stroma in relation with clinicopathological features in CRC patients. METHOD Immunohistochemical staining of LOX proteins was performed on tissue microarrays from colorectal tumor samples taken from resection specimens. LOX expression was quantified in tumor cells and stroma. The correlation between the expression of LOX and clinicopathological parameters was analyzed. RESULTS A positive correlation was observed between peritumoral stromal LOX expression and LOX expression in the tumor epithelium. High expression of LOX in tumor cells was significantly associated with poorer progression-free survival (PFS) among patients. Low tumor budding was observed in tumors with low stromal LOX expression. CONCLUSION The current study indicates that LOX may be an important contributor to CRC progression. The findings of this series, in which LOX expression correlated with tumor budding and survival, support a contribution for LOX to EMT and metastasis. Furthermore, LOX expression in both the tumor cell and stromal compartment may add information to improve prognosis in CRC management. These findings, however, have to be validated in further studies, as does also the investigation of LOX as a potential therapeutic target in CRC.
Collapse
Affiliation(s)
| | - Seda Kahraman
- Department of Medical Oncology, Ankara Yıldırım Beyazıt University, Ankara, Turkey
| | - Doğukan Çanakçı
- Ankara Yıldırım Beyazıt University Faculty of Medicine, Ankara, Turkey
| | - Mustafa Tahtacı
- Department of Gastroenterology, Ankara Yıldırım Beyazıt University Faculty of Medicine, Ankara, Turkey
| | - Ayşegül Aksoy Altınboğa
- Department of Pathology, Ankara Yıldırım Beyazıt University Faculty of Medicine, Ankara, Turkey
| | - Hayriye Tatlı Doğan
- Department of Pathology, Ankara Yıldırım Beyazıt University Faculty of Medicine, Ankara, Turkey.
| |
Collapse
|
3
|
Zhou Y, Jiang Z, Cao L, Yang J. The role of various collagen types in tumor biology: a review. Front Oncol 2025; 15:1549797. [PMID: 40110201 PMCID: PMC11919678 DOI: 10.3389/fonc.2025.1549797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
Collagen comprises approximately 30% of the body's protein content and is essential for maintaining the structural integrity, support, and strength of the skin, muscles, bones, and connective tissues. Recent research has further elucidated its role in various aspects of tumor biology, including tumorigenesis, invasion, migration, drug resistance, and recurrence. Furthermore, collagen is involved in prognostic assessments, the evaluation of therapeutic efficacy, immunoregulation, and the identification of potential treatment targets in oncology. This review examines a range of tumor types, including lung, gastric, breast, melanoma, and colorectal cancers, among others. Our objective is to differentiate these tumors based on the specific types of collagen present and to analyze the roles of various collagen types in tumor development, progression, prognosis, and their potential as therapeutic targets.
Collapse
Affiliation(s)
- Yuchuan Zhou
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Zhonghui Jiang
- Thoracic Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Lu Cao
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jianquan Yang
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
4
|
Gupta A, Choudhary P, Singh S. Identification and targeting of metastatic biomarkers for hepatocellular carcinoma therapeutics using small molecules library of curcumin analogues. Mol Divers 2025; 29:503-517. [PMID: 38689175 DOI: 10.1007/s11030-024-10871-3] [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: 02/08/2024] [Accepted: 04/03/2024] [Indexed: 05/02/2024]
Abstract
The understanding of the molecular basis of complex diseases like hepatocellular carcinoma (HCC) needs large datasets of multiple genes and proteins involved in different phenomenon of its development. This study focuses on the molecular basis of HCC and the development of therapeutic strategies. We analyzed a dataset of 5475 genes (Homo sapiens) involved in HCC hallmarks, involving comprehensive data on multiple genes and frequently mutated genes. As HCC is characterized by metastasis, angiogenesis, and oxidative stress, exploration of genes associated with them has been targeted. Through gene ontology, functional characterization, and pathway enrichment analysis, we identified target proteins such as Lysyl oxidase, Survivin, Cofilin, and Cathepsin B. A library of curcumin analogs was used to target these proteins. Tetrahrydrocurcumin showed promising binding affinities for all four proteins, suggesting its potential as an inhibitor against these proteins for HCC therapy.
Collapse
Affiliation(s)
- Ayushi Gupta
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, Devghat, Jhalwa, Prayagraj, 211015, Uttar Pradesh, India
| | - Princy Choudhary
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, Devghat, Jhalwa, Prayagraj, 211015, Uttar Pradesh, India
| | - Sangeeta Singh
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, Devghat, Jhalwa, Prayagraj, 211015, Uttar Pradesh, India.
| |
Collapse
|
5
|
Kamiya T, Ishii K, Ozawa K, Hara H. Bortezomib suppresses TGF-β1-mediated LOXL4 reduction through the inhibition of MEK/ERK pathways in MDA-MB-231 cells. J Recept Signal Transduct Res 2025; 45:73-82. [PMID: 39862152 DOI: 10.1080/10799893.2025.2455594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 01/06/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025]
Abstract
Lysyl oxidase (LOX), a copper-containing secretory oxidase, plays a key role in the regulation of extracellular stiffness through cross-linking with collagen and elastin. Among the LOX family of enzymes, LOX-like 4 (LOXL4) exhibits pro-tumor and anti-tumor properties; therefore, the functional role of LOXL4 in tumor progression is still under investigation. Here, we first determined that transforming growth factor-β1 (TGF-β1) significantly decreased LOXL4 expression in human breast cancer MDA-MB-231 cells, which suggested that decreased LOXL4 may participate in tumor progression. In this study, we also investigated how TGF-β1 decreases LOXL4 expression. TGF-β1-induced intracellular reactive oxygen species (ROS) played a role in LOXL4 protein expression but had no effect on LOXL4 mRNA levels. The proteasomal inhibitor, bortezomib, significantly suppressed TGF-β1-mediated LOXL4 reduction, which indicated that TGF-β1 facilitates LOXL4 proteasomal degradation. Furthermore, bortezomib inhibited TGF-β1-induced MEK/ERK pathways which are involved in LOXL4 reduction and TGF-β1-mediated cell migration. Finally, we also determined the potential role of N-glycosylation in LOXL4 secretion. We found that the dysregulation of N-glycosylation may be involved in the reduction in LOXL4 secretion. Overall, bortezomib is expected to inhibit TNBC progression by inhibiting both the MEK/ERK and proteasomal degradation pathways, which regulate LOXL4 expression.
Collapse
Affiliation(s)
- Tetsuro Kamiya
- Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Kana Ishii
- Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Kiyomi Ozawa
- Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Hirokazu Hara
- Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
6
|
Wang Z, Wang W, Luo Q, Song G. High matrix stiffness accelerates migration of hepatocellular carcinoma cells through the integrin β1-Plectin-F-actin axis. BMC Biol 2025; 23:8. [PMID: 39789506 PMCID: PMC11721467 DOI: 10.1186/s12915-025-02113-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Abundant research indicates that increased extracellular matrix (ECM) stiffness significantly enhances the malignant characteristics of hepatocellular carcinoma (HCC) cells. Plectin, an essential cytoskeletal linker protein, has recently emerged as a promoter of cancer progression, particularly in the context of cancer cell invasion and metastasis. However, the responsiveness of plectin to changes in ECM stiffness and its impact on HCC progression remain unclear. In this study, we aimed to investigate whether plectin responds to variations in ECM stiffness and to explore its involved molecular mechanisms in regulating HCC cell migration. RESULTS Our results showed that, when compared with control group (7 kPa), high ECM stiffness (53 kPa) boosts HCC cell migration by upregulating plectin and integrin β1 expression and increasing F-actin polymerization. Knockdown of integrin β1 negated the high stiffness-upregulated plectin expression. Furthermore, reducing either plectin or integrin β1 levels, or using latrunculin A, effectively prevented the high ECM stiffness-induced F-actin polymerization and HCC cell migration. CONCLUSIONS These findings demonstrate that integrin β1-plectin-F-actin axis is necessary for high matrix stiffness-driven migration of HCC cells, and provide evidence for the critical role of plectin in mechanotransduction in HCC cells.
Collapse
Affiliation(s)
- Zhihui Wang
- College of Bioengineering, Chongqing University, Chongqing, 400030, China
- Key Laboratory of Biorheological Science & Technology, Ministry of Education, Chongqing University, Chongqing, 400030, China
| | - Wenbin Wang
- College of Bioengineering, Chongqing University, Chongqing, 400030, China
- Key Laboratory of Biorheological Science & Technology, Ministry of Education, Chongqing University, Chongqing, 400030, China
| | - Qing Luo
- College of Bioengineering, Chongqing University, Chongqing, 400030, China
- Key Laboratory of Biorheological Science & Technology, Ministry of Education, Chongqing University, Chongqing, 400030, China
| | - Guanbin Song
- College of Bioengineering, Chongqing University, Chongqing, 400030, China.
- Key Laboratory of Biorheological Science & Technology, Ministry of Education, Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
7
|
Lu Y, Li H, Chen M, Lin Y, Zhang X. LOX-induced tubulointerstitial fibrosis via the TGF-β/LOX/Snail axis in diabetic mice. J Transl Med 2025; 23:35. [PMID: 39789539 PMCID: PMC11716213 DOI: 10.1186/s12967-024-06056-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 12/24/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND The partial epithelial-mesenchymal transition (EMT) is emerging as a significant mechanism in diabetic nephropathy (DN). LOX is a copper amine oxidase conventionally thought to act by crosslinking collagen. However, the role of LOX in partial EMT and fibrotic progression in diabetic nephropathy has not been investigated experimentally. METHODS The bulk RNA sequencing and single-nuclei RNA sequencing (snRNA-seq) analysis were explored to find the role of LOX in diabetic nephropathy. We then investigated the partial EMT and the possible signaling pathway of LOX, both in vivo and in vitro by LOX inhibition experiments in diabetic mice and HK-2 cells. Besides, we further assessed kidney fibrosis and renal function. RESULTS LOX expression was elevated in kidneys of diabetic mice. Additionally, snRNA-seq results indicated that LOX expression was higher in partial epithelial-mesenchymal transition proximal tubular (PemtPT) epithelial cells. Moreover, we found that increased LOX prompted partial EMT of renal tubular epithelial cells (RTECs) by modulating the transcription factor Snail both in vivo and in vitro. Remarkably, inhibition of LOX effectively mitigated the partial EMT of RTECs in diabetic mice, thereby attenuating kidney fibrosis and enhancing renal function. Additionally, we identified the TGF-β signaling pathway as an upstream regulator of LOX, and inhibiting LOX partially reversed the partial EMT program in HK-2 cells induced by the TGF-β signaling pathway. CONCLUSIONS Hyperglycemia induces partial EMT of RTECs via the TGF-β/LOX/Snail axis, thereby contributing to diabetic kidney fibrosis. Inhibiting LOX can effectively reverse the partial EMT of RTECs, diminish diabetic kidney fibrosis, and improve renal function.
Collapse
Affiliation(s)
- Yicheng Lu
- School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Heyangzi Li
- Department of Pathology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Mohan Chen
- School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yicheng Lin
- Xiangya School of Medicine, Central South University, Changsha, 410083, China
| | - Xiaoming Zhang
- Department of Basic Medical Sciences, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
8
|
Mezentsev A, Durymanov M, Makarov VA. A Comprehensive Review of Protein Biomarkers for Invasive Lung Cancer. Curr Oncol 2024; 31:4818-4854. [PMID: 39329988 PMCID: PMC11431409 DOI: 10.3390/curroncol31090360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/16/2024] [Accepted: 08/22/2024] [Indexed: 09/28/2024] Open
Abstract
Invasion and metastasis are important hallmarks of lung cancer, and affect patients' survival. Early diagnostics of metastatic potential are important for treatment management. Recent findings suggest that the transition to an invasive phenotype causes changes in the expression of 700-800 genes. In this context, the biomarkers restricted to the specific type of cancer, like lung cancer, are often overlooked. Some well-known protein biomarkers correlate with the progression of the disease and the immunogenicity of the tumor. Most of these biomarkers are not exclusive to lung cancer because of their significant role in tumorigenesis. The dysregulation of others does not necessarily indicate cell invasiveness, as they play an active role in cell division. Clinical studies of lung cancer use protein biomarkers to assess the invasiveness of cancer cells for therapeutic purposes. However, there is still a need to discover new biomarkers for lung cancer. In the future, minimally invasive techniques, such as blood or saliva analyses, may be sufficient for this purpose. Many researchers suggest unconventional biomarkers, like circulating nucleic acids, exosomal proteins, and autoantibodies. This review paper aims to discuss the advantages and limitations of protein biomarkers of invasiveness in lung cancer, to assess their prognostic value, and propose novel biomarker candidates.
Collapse
Affiliation(s)
- Alexandre Mezentsev
- Medical Informatics Laboratory, Yaroslav-the-Wise Novgorod State University, 173003 Veliky Novgorod, Russia; (M.D.); (V.A.M.)
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia
| | - Mikhail Durymanov
- Medical Informatics Laboratory, Yaroslav-the-Wise Novgorod State University, 173003 Veliky Novgorod, Russia; (M.D.); (V.A.M.)
| | - Vladimir A. Makarov
- Medical Informatics Laboratory, Yaroslav-the-Wise Novgorod State University, 173003 Veliky Novgorod, Russia; (M.D.); (V.A.M.)
| |
Collapse
|
9
|
Yuan GQ, Zhang G, Nie Q, Wang Z, Gao HZ, Jin GS, Zheng ZQ. Lysyl oxidase-like 1 predicts the prognosis of patients with primary glioblastoma and promotes tumor invasion via EMT pathway. PeerJ 2024; 12:e17579. [PMID: 38978755 PMCID: PMC11229686 DOI: 10.7717/peerj.17579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 05/25/2024] [Indexed: 07/10/2024] Open
Abstract
Background Lysyl oxidase enzymes (LOXs), as extracellular matrix (ECM) protein regulators, play vital roles in tumor progression by remodeling the tumor microenvironment. However, their roles in glioblastoma (GBM) have not been fully elucidated. Methods The genetic alterations and prognostic value of LOXs were investigated via cBioPortal. The correlations between LOXs and biological functions/molecular tumor subtypes were explored in The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA). After Kaplan‒Meier and Cox survival analyses, a Loxl1-based nomogram and prognostic risk score model (PRSM) were constructed and evaluated by time-dependent receiver operating characteristic curves, calibration curves, and decision curve analyses. Tumor enrichment pathways and immune infiltrates were explored by single-cell RNA sequencing and TIMER. Loxl1-related changes in tumor viability/proliferation and invasion were further validated by CCK-8, western blot, wound healing, and Transwell invasion assays. Results GBM patients with altered LOXs had poor survival. Upregulated LOXs were found in IDH1-wildtype and mesenchymal (not Loxl1) GBM subtypes, promoting ECM receptor interactions in GBM. The Loxl1-based nomogram and the PRSM showed high accuracy, reliability, and net clinical benefits. Loxl1 expression was related to tumor invasion and immune infiltration (B cells, neutrophils, and dendritic cells). Loxl1 knockdown suppressed GBM cell proliferation and invasion by inhibiting the EMT pathway (through the downregulation of N-cadherin/Vimentin/Snai1 and the upregulation of E-cadherin). Conclusion The Loxl1-based nomogram and PRSM were stable and individualized for assessing GBM patient prognosis, and the invasive role of Loxl1 could provide a promising therapeutic strategy.
Collapse
Affiliation(s)
- Gui-Qiang Yuan
- Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Capital Medical University, Beijing, China
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Guoguo Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qianqian Nie
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hong-Zhi Gao
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Gui-Shan Jin
- Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Capital Medical University, Beijing, China
| | - Zong-Qing Zheng
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- Department of Neurosurgery, Neurosurgery Research Institute & Binhai Branch of National Regional Medical Center, The First Affiliated Hospital, Fujian Medical University, Fujian, Fuzhou, China
| |
Collapse
|
10
|
Škarková A, Bizzarri M, Janoštiak R, Mašek J, Rosel D, Brábek J. Educate, not kill: treating cancer without triggering its defenses. Trends Mol Med 2024; 30:673-685. [PMID: 38658206 DOI: 10.1016/j.molmed.2024.04.003] [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: 02/19/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/26/2024]
Abstract
Traditionally, anticancer therapies focus on restraining uncontrolled proliferation. However, these cytotoxic therapies expose cancer cells to direct killing, instigating the process of natural selection favoring survival of resistant cells that become the foundation for tumor progression and therapy failure. Recognizing this phenomenon has prompted the development of alternative therapeutic strategies. Here we propose strategies targeting cancer hallmarks beyond proliferation, aiming at re-educating cancer cells towards a less malignant phenotype. These strategies include controlling cell dormancy, transdifferentiation therapy, normalizing the cancer microenvironment, and using migrastatic therapy. Adaptive resistance to these educative strategies does not confer a direct proliferative advantage to resistant cells, as non-resistant cells are not subject to eradication, thereby delaying or preventing the development of therapy-resistant tumors.
Collapse
Affiliation(s)
- Aneta Škarková
- Department of Cell Biology, BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic
| | - Mariano Bizzarri
- System Biology Group Laboratory, Sapienza University, Rome, Italy
| | - Radoslav Janoštiak
- First Faculty of Medicine, BIOCEV, Charles University, Vestec, Czech Republic
| | - Jan Mašek
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Daniel Rosel
- Department of Cell Biology, BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic.
| | - Jan Brábek
- Department of Cell Biology, BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic.
| |
Collapse
|
11
|
Chuang TD, Ton N, Rysling S, Khorram O. In Vivo Effects of Bay 11-7082 on Fibroid Growth and Gene Expression: A Preclinical Study. Cells 2024; 13:1091. [PMID: 38994944 PMCID: PMC11240737 DOI: 10.3390/cells13131091] [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: 05/16/2024] [Revised: 06/18/2024] [Accepted: 06/21/2024] [Indexed: 07/13/2024] Open
Abstract
Current medical therapies for fibroids have major limitations due to their hypoestrogenic side effects. Based on our previous work showing the activation of NF-kB in fibroids, we hypothesized that inhibiting NF-kB in vivo would result in the shrinkage of tumors and reduced inflammation. Fibroid xenografts were implanted in SCID mice and treated daily with Bay 11-7082 (Bay) or vehicle for two months. Bay treatment led to a 50% reduction in tumor weight. RNAseq revealed decreased expression of genes related to cell proliferation, inflammation, extracellular matrix (ECM) composition, and growth factor expression. Validation through qRT-PCR, Western blotting, ELISA, and immunohistochemistry (IHC) confirmed these findings. Bay treatment reduced mRNA expression of cell cycle regulators (CCND1, E2F1, and CKS2), inflammatory markers (SPARC, TDO2, MYD88, TLR3, TLR6, IL6, TNFα, TNFRSF11A, and IL1β), ECM remodelers (COL3A1, FN1, LOX, and TGFβ3), growth factors (PRL, PDGFA, and VEGFC), progesterone receptor, and miR-29c and miR-200c. Collagen levels were reduced in Bay-treated xenografts. Western blotting and IHC showed decreased protein abundance in certain ECM components and inflammatory markers, but not cleaved caspase three. Ki67, CCND1, and E2F1 expression decreased with Bay treatment. This preclinical study suggests NF-kB inhibition as an effective fibroid treatment, suppressing genes involved in proliferation, inflammation, and ECM remodeling.
Collapse
Affiliation(s)
- Tsai-Der Chuang
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (T.-D.C.); (N.T.); (S.R.)
| | - Nhu Ton
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (T.-D.C.); (N.T.); (S.R.)
| | - Shawn Rysling
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (T.-D.C.); (N.T.); (S.R.)
| | - Omid Khorram
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (T.-D.C.); (N.T.); (S.R.)
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California, Los Angeles, CA 90024, USA
| |
Collapse
|
12
|
Lin X, Yang P, Wang M, Huang X, Wang B, Chen C, Xu A, Cai J, Khan M, Liu S, Lin J. Dissecting gastric cancer heterogeneity and exploring therapeutic strategies using bulk and single-cell transcriptomic analysis and experimental validation of tumor microenvironment and metabolic interplay. Front Pharmacol 2024; 15:1355269. [PMID: 38962317 PMCID: PMC11220201 DOI: 10.3389/fphar.2024.1355269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/15/2024] [Indexed: 07/05/2024] Open
Abstract
Gastric cancer, the fifth most prevalent cancer worldwide, is often diagnosed in advanced stages with limited treatment options. Examining the tumor microenvironment (TME) and its metabolic reprogramming can provide insights for better diagnosis and treatment. This study investigates the link between TME factors and metabolic activity in gastric cancer using bulk and single-cell RNA-sequencing data. We identified two molecular subtypes in gastric cancer by analyzing the distinct expression patterns of 81 prognostic genes related to the TME and metabolism, which exhibited significant protein-level interactions. The high-risk subtype had increased stromal content, fibroblast and M2 macrophage infiltration, elevated glycosaminoglycans/glycosphingolipids biosynthesis, and fat metabolism, along with advanced clinicopathological features. It also exhibited low mutation rates and microsatellite instability, associating it with the mesenchymal phenotype. In contrast, the low-risk group showed higher tumor content and upregulated protein and sugar metabolism. We identified a 15-gene prognostic signature representing these characteristics, including CPVL, KYNU, CD36, and GPX3, strongly correlated with M2 macrophages, validated through single-cell analysis and an internal cohort. Despite resistance to immunotherapy, the high-risk group showed sensitivity to molecular targeted agents directed at IGF-1R (BMS-754807) and the PI3K-mTOR pathways (AZD8186, AZD8055). We experimentally validated these promising drugs for their inhibitory effects on MKN45 and MKN28 gastric cells. This study unveils the intricate interplay between TME and metabolic pathways in gastric cancer, offering potential for enhanced diagnosis, patient stratification, and personalized treatment. Understanding molecular features in each subtype enriches our comprehension of gastric cancer heterogeneity and potential therapeutic targets.
Collapse
Affiliation(s)
- XianTao Lin
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Ping Yang
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - MingKun Wang
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Xiuting Huang
- Department of Radiotherapy, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Baiyao Wang
- Department of Radiotherapy, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Chengcong Chen
- Department of Radiotherapy, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Anan Xu
- Department of Radiotherapy, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Jiazuo Cai
- Department of Radiotherapy, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Muhammad Khan
- Department of Radiotherapy, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Sha Liu
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Jie Lin
- Department of Radiotherapy, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
13
|
Seifi Z, Khazaei M, Cheraghali D, Rezakhani L. Decellularized tissues as platforms for digestive system cancer models. Heliyon 2024; 10:e31589. [PMID: 38845895 PMCID: PMC11153114 DOI: 10.1016/j.heliyon.2024.e31589] [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: 09/20/2023] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/09/2024] Open
Abstract
The extracellular matrix (ECM) is a multifunctional network of macromolecules that regulate various cellular functions and physically support the tissues. Besides physiological conditions, the ECM also changes during pathological conditions such as cancer. As tumor cells proliferate, notable changes occur in the quantity and makeup of the surrounding ECM. Therefore, the role of this noncellular component of tissues in studies of tumor microenvironments should be considered. So far, many attempts have been made to create 2-dimensional (2D) or 3-dimensional (3D) models that can replicate the intricate connections within the tumor microenvironment. Decellularized tissues are proper scaffolds that imitate the complex nature of native ECM. This review aims to summarize 3D models of digestive system cancers based on decellularized ECMs. These ECM-based scaffolds will enable us to study the interactive communication between cells and their surrounding environment which brings new potential for a better understanding of the pathophysiology of cancer.
Collapse
Affiliation(s)
- Zahra Seifi
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mozafar Khazaei
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Danial Cheraghali
- Department of Mechanical Engineering, New Jersey Institute of Technology, NJ, USA
| | - Leila Rezakhani
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
14
|
Nakajima K, Suzuki M, Kawashima I, Koshiisi M, Kumagai T, Yamamoto T, Tanaka M, Kirito K. The chaperone protein GRP78 released from MPN cells increases the expression of lysyl oxidase in a human stromal cell line. Leuk Res 2023; 134:107389. [PMID: 37757654 DOI: 10.1016/j.leukres.2023.107389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 08/30/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023]
Abstract
Impaired function of the endoplasmic stress (ER) response causes numerous pathological conditions, including tissue fibrosis. In the present study, we aimed to determine the pathological role of ER stress response systems in myeloproliferative neoplasms (MPNs). We found increased expression of the chaperone protein glucose-regulated protein (GRP) 78, a central regulator of ER stress, in megakaryocytes from primary myelofibrosis or postessential thrombocythemia myelofibrosis patients. GRP78 was overexpressed in JAK2V617F-harboring cell lines; however, inhibitors of ER stress did not affect the expression levels of GRP78. In contrast, ruxolitinib, a well-known inhibitor of JAK2V617F, clearly blocked GRP78 expression in these cells through downregulation of transcription factor 4 (ATF4). Interestingly, GRP78 was secreted from HEL and SET-2 cells into culture media. Coculture of these cells with HS-5 cells, a human bone marrow stroma-derived cell line, induced enhanced expression of lysyl oxidase (LOX), which mediates cross-linking of collagen fibers and induces tissue fibrosis, in HS-5 cells. An anti-GRP78 neutralizing antibody abrogated LOX elevation; in contrast, recombinant GRP78 protein induced LOX protein expression in HS-5 cells. Our observations suggest that the oncogenic protein JAK2V617F induces overexpression and release of GRP78, which may induce a fibrotic phenotype in surrounding bone marrow stromal cells.
Collapse
Affiliation(s)
- Kei Nakajima
- Department of Hematology/Oncology, University of Yamanashi, Japan
| | - Megumi Suzuki
- Department of Hematology/Oncology, University of Yamanashi, Japan
| | - Ichiro Kawashima
- Department of Hematology/Oncology, University of Yamanashi, Japan
| | - Megumi Koshiisi
- Department of Hematology/Oncology, University of Yamanashi, Japan
| | - Takuma Kumagai
- Department of Hematology/Oncology, University of Yamanashi, Japan
| | - Takeo Yamamoto
- Department of Hematology/Oncology, University of Yamanashi, Japan
| | - Masaru Tanaka
- Department of Hematology/Oncology, University of Yamanashi, Japan
| | - Keita Kirito
- Department of Hematology/Oncology, University of Yamanashi, Japan.
| |
Collapse
|
15
|
Zhou H, Lu D, Yu D, Luo C, Xie K, Ma H, Li S, Liang J, Wei F, Chen L, Luo D, Wang W, Wei J. Pan-cancer analysis of the oncogenic role of the core osteosarcoma gene VCAN in human tumors. Am J Transl Res 2023; 15:5556-5573. [PMID: 37854213 PMCID: PMC10579017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 08/30/2023] [Indexed: 10/20/2023]
Abstract
OBJECTIVE Versican (VCAN), a member of the multifunctional glycoprotein family, is involved in various aspects of cancer progression. However, the role of VCAN in diverse cancers remains poorly defined. This research aimed to investigate the correlation between VCAN expression and the oncogenic role, as well as visualize its prognostic landscape in pan-cancer. METHODS Raw data in regard to VCAN expression in cancer patients were acquired from GEO GeneChip public database in NCBI. Besides, we selected microarray data GSE16088 for analysis. We retrieved the genes associated with osteosarcoma (OS) from the OMIM database and identified their intersection with the core module. VCAN was suggested to be a potential marker gene for OS. Subsequently, we conducted Gene Set Enrichment Analysis (GSEA) to explore gene functional enrichment. Moreover, we performed pan-cancer analysis on VCAN to gain a comprehensive understanding of its implications across various cancer types. RESULTS The VCAN expression in the tumor tissue was higher than that in normal tissue. Elevated expression of VCAN was associated with high the tumor stage and poor long-term survival. There was a significant positive correlation between VCAN and cancer fibroblasts in all pan cancers. Moreover, FBN1 was the intersection gene of VCAN-related genes and linker genes. ANTXR1, COL5A2, CSGALNACT2, and SPARC were the target genes of VCAN genes. GSEA analysis showed that VCAN was mainly enriched in the extracellular matrix (ECM) signaling pathway. CONCLUSION VCAN can be used as a marker molecule for the early diagnosis of OS and holds significance as a molecule in cases of OS with distant metastasis. The ECM signaling pathway may be a core pathway in OS development and distant metastasis. These findings shed new light on therapeutics of cancers.
Collapse
Affiliation(s)
- Haidong Zhou
- Graduate School of Youjiang Medical University for NationalitiesBaise 533000, Guangxi, China
- Department of Sports Medicine, Baidong Hospital Affiliated to Youjiang Medical College for NationalitiesBaise 533000, Guangxi, China
| | - Dinggui Lu
- Department of Trauma Orthopedics, Baidong Hospital, Youjiang Medical College for NationalitiesBaise 533000, Guangxi, China
| | - Dianbo Yu
- Department of Sports Medicine, Baidong Hospital Affiliated to Youjiang Medical College for NationalitiesBaise 533000, Guangxi, China
| | - Changtai Luo
- Graduate School of Youjiang Medical University for NationalitiesBaise 533000, Guangxi, China
| | - Kangqi Xie
- Department of Sports Medicine, Baidong Hospital Affiliated to Youjiang Medical College for NationalitiesBaise 533000, Guangxi, China
| | - Huade Ma
- Department of Sports Medicine, Baidong Hospital Affiliated to Youjiang Medical College for NationalitiesBaise 533000, Guangxi, China
| | - Shanlang Li
- Department of Sports Medicine, Baidong Hospital Affiliated to Youjiang Medical College for NationalitiesBaise 533000, Guangxi, China
| | - Jiyun Liang
- Graduate School of Youjiang Medical University for NationalitiesBaise 533000, Guangxi, China
| | - Fengxu Wei
- Graduate School of Youjiang Medical University for NationalitiesBaise 533000, Guangxi, China
| | - Luchang Chen
- Graduate School of Youjiang Medical University for NationalitiesBaise 533000, Guangxi, China
| | - Dong Luo
- Graduate School of Youjiang Medical University for NationalitiesBaise 533000, Guangxi, China
| | - Wei Wang
- Graduate School of Youjiang Medical University for NationalitiesBaise 533000, Guangxi, China
| | - Jihua Wei
- Department of Sports Medicine, Baidong Hospital Affiliated to Youjiang Medical College for NationalitiesBaise 533000, Guangxi, China
| |
Collapse
|
16
|
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
|
17
|
Zhang S, Regan K, Najera J, Grinstaff MW, Datta M, Nia HT. The peritumor microenvironment: physics and immunity. Trends Cancer 2023; 9:609-623. [PMID: 37156677 PMCID: PMC10523902 DOI: 10.1016/j.trecan.2023.04.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/05/2023] [Accepted: 04/11/2023] [Indexed: 05/10/2023]
Abstract
Cancer initiation and progression drastically alter the microenvironment at the interface between healthy and malignant tissue. This site, termed the peritumor, bears unique physical and immune attributes that together further promote tumor progression through interconnected mechanical signaling and immune activity. In this review, we describe the distinct physical features of the peritumoral microenvironment and link their relationship to immune responses. The peritumor is a region rich in biomarkers and therapeutic targets and thus is a key focus for future cancer research as well as clinical outlooks, particularly to understand and overcome novel mechanisms of immunotherapy resistance.
Collapse
Affiliation(s)
- Sue Zhang
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Kathryn Regan
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Julian Najera
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - Mark W Grinstaff
- Department of Biomedical Engineering, Boston University, Boston, MA, USA; Department of Chemistry, Boston University, Boston, MA, USA
| | - Meenal Datta
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, USA.
| | - Hadi T Nia
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
| |
Collapse
|
18
|
Radić J, Kožik B, Nikolić I, Kolarov-Bjelobrk I, Vasiljević T, Vranjković B, Despotović S. Multiple Roles of LOXL2 in the Progression of Hepatocellular Carcinoma and Its Potential for Therapeutic Targeting. Int J Mol Sci 2023; 24:11745. [PMID: 37511503 PMCID: PMC10380739 DOI: 10.3390/ijms241411745] [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: 06/10/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
LOXL2, a copper-dependent amine oxidase, has emerged as a promising therapeutic target in hepatocellular carcinoma (HCC). Increased LOXL2 expression in HCC has been linked with an aggressive phenotype and represents a poor prognostic factor. Here, we focus on the mechanisms through which LOXL2 orchestrates multiple oncogenic functions in HCC development. We performed a review of the current knowledge on the roles LOXL2 performs in the modulation of the HCC tumor microenvironment, formation of premetastatic niches, and epithelial-mesenchymal transition. We also highlighted the complex interplay between LOXL2 and hypoxia, angiogenesis, and vasculogenic mimicry in HCC. At the end of the review, we summarize the current LOXL2 inhibitors and discuss their potential in HCC precision treatment.
Collapse
Affiliation(s)
- Jelena Radić
- Faculty of Medicine, University of Novi Sad, 21137 Novi Sad, Serbia
- Department of Medical Oncology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Serbia
| | - Bojana Kožik
- Laboratory for Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 11100 Belgrade, Serbia
| | - Ivan Nikolić
- Faculty of Medicine, University of Novi Sad, 21137 Novi Sad, Serbia
- Department of Medical Oncology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Serbia
| | - Ivana Kolarov-Bjelobrk
- Faculty of Medicine, University of Novi Sad, 21137 Novi Sad, Serbia
- Department of Medical Oncology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Serbia
| | - Tijana Vasiljević
- Faculty of Medicine, University of Novi Sad, 21137 Novi Sad, Serbia
- Department of Pathology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Serbia
| | - Bojana Vranjković
- Department of Medical Oncology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Serbia
| | - Sanja Despotović
- Institute of Histology and Embryology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
19
|
Zhou C, Yang J, Liu T, Jia R, Yang L, Sun P, Zhao W. Copper metabolism and hepatocellular carcinoma: current insights. Front Oncol 2023; 13:1186659. [PMID: 37476384 PMCID: PMC10355993 DOI: 10.3389/fonc.2023.1186659] [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: 03/15/2023] [Accepted: 06/21/2023] [Indexed: 07/22/2023] Open
Abstract
Copper is an essential trace element that acts as a cofactor in various enzyme active sites in the human body. It participates in numerous life activities, including lipid metabolism, energy metabolism, and neurotransmitter synthesis. The proposal of "Cuproptosis" has made copper metabolism-related pathways a research hotspot in the field of tumor therapy, which has attracted great attention. This review discusses the biological processes of copper uptake, transport, and storage in human cells. It highlights the mechanisms by which copper metabolism affects hepatocellular carcinogenesis and metastasis, including autophagy, apoptosis, vascular invasion, cuproptosis, and ferroptosis. Additionally, it summarizes the current clinical applications of copper metabolism-related drugs in antitumor therapy.
Collapse
Affiliation(s)
- Cheng Zhou
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jinqiu Yang
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Tong Liu
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ran Jia
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Lin Yang
- Department of Hepatobiliary Surgery, Xianyang Central Hospital Affiliated to Shaanxi University of Chinese Medicine, Xianyang, China
| | - Pengfei Sun
- Department of Orthopaedics, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Wenxia Zhao
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
20
|
Kuziel G, Moore BN, Arendt LM. Obesity and Fibrosis: Setting the Stage for Breast Cancer. Cancers (Basel) 2023; 15:cancers15112929. [PMID: 37296891 DOI: 10.3390/cancers15112929] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Obesity is a rising health concern and is linked to a worsened breast cancer prognosis. Tumor desmoplasia, which is characterized by elevated numbers of cancer-associated fibroblasts and the deposition of fibrillar collagens within the stroma, may contribute to the aggressive clinical behavior of breast cancer in obesity. A major component of the breast is adipose tissue, and fibrotic changes in adipose tissue due to obesity may contribute to breast cancer development and the biology of the resulting tumors. Adipose tissue fibrosis is a consequence of obesity that has multiple sources. Adipocytes and adipose-derived stromal cells secrete extracellular matrix composed of collagen family members and matricellular proteins that are altered by obesity. Adipose tissue also becomes a site of chronic, macrophage-driven inflammation. Macrophages exist as a diverse population within obese adipose tissue and mediate the development of fibrosis through the secretion of growth factors and matricellular proteins and interactions with other stromal cells. While weight loss is recommended to resolve obesity, the long-term effects of weight loss on adipose tissue fibrosis and inflammation within breast tissue are less clear. Increased fibrosis within breast tissue may increase the risk for tumor development as well as promote characteristics associated with tumor aggressiveness.
Collapse
Affiliation(s)
- Genevra Kuziel
- Cancer Biology Graduate Program, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA
| | - Brittney N Moore
- Department of Comparative Biosciences, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI 53706, USA
| | - Lisa M Arendt
- Cancer Biology Graduate Program, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA
- Department of Comparative Biosciences, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI 53706, USA
| |
Collapse
|
21
|
Bruni S, Mercogliano MF, Mauro FL, Cordo Russo RI, Schillaci R. Cancer immune exclusion: breaking the barricade for a successful immunotherapy. Front Oncol 2023; 13:1135456. [PMID: 37284199 PMCID: PMC10239871 DOI: 10.3389/fonc.2023.1135456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 05/10/2023] [Indexed: 06/08/2023] Open
Abstract
Immunotherapy has changed the course of cancer treatment. The initial steps were made through tumor-specific antibodies that guided the setup of an antitumor immune response. A new and successful generation of antibodies are designed to target immune checkpoint molecules aimed to reinvigorate the antitumor immune response. The cellular counterpart is the adoptive cell therapy, where specific immune cells are expanded or engineered to target cancer cells. In all cases, the key for achieving positive clinical resolutions rests upon the access of immune cells to the tumor. In this review, we focus on how the tumor microenvironment architecture, including stromal cells, immunosuppressive cells and extracellular matrix, protects tumor cells from an immune attack leading to immunotherapy resistance, and on the available strategies to tackle immune evasion.
Collapse
|
22
|
Dzobo K, Dandara C. The Extracellular Matrix: Its Composition, Function, Remodeling, and Role in Tumorigenesis. Biomimetics (Basel) 2023; 8:146. [PMID: 37092398 PMCID: PMC10123695 DOI: 10.3390/biomimetics8020146] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/25/2023] Open
Abstract
The extracellular matrix (ECM) is a ubiquitous member of the body and is key to the maintenance of tissue and organ integrity. Initially thought to be a bystander in many cellular processes, the extracellular matrix has been shown to have diverse components that regulate and activate many cellular processes and ultimately influence cell phenotype. Importantly, the ECM's composition, architecture, and stiffness/elasticity influence cellular phenotypes. Under normal conditions and during development, the synthesized ECM constantly undergoes degradation and remodeling processes via the action of matrix proteases that maintain tissue homeostasis. In many pathological conditions including fibrosis and cancer, ECM synthesis, remodeling, and degradation is dysregulated, causing its integrity to be altered. Both physical and chemical cues from the ECM are sensed via receptors including integrins and play key roles in driving cellular proliferation and differentiation and in the progression of various diseases such as cancers. Advances in 'omics' technologies have seen an increase in studies focusing on bidirectional cell-matrix interactions, and here, we highlight the emerging knowledge on the role played by the ECM during normal development and in pathological conditions. This review summarizes current ECM-targeted therapies that can modify ECM tumors to overcome drug resistance and better cancer treatment.
Collapse
Affiliation(s)
- Kevin Dzobo
- Medical Research Council, SA Wound Healing Unit, Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Groote Schuur Hospital, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa
| | - Collet Dandara
- Division of Human Genetics and Institute of Infectious Disease and Molecular Medicine, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa
- The South African Medical Research Council-UCT Platform for Pharmacogenomics Research and Translation, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa
| |
Collapse
|
23
|
Wang S, Zheng C, Huang Y, He X. Betel quid may stimulate oral submucous fibrosis by inducing increased mitochondrial reactive oxygen species generation via copper overload. Med Hypotheses 2023. [DOI: 10.1016/j.mehy.2023.111067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
24
|
Transcriptome-Based Traits of Radioresistant Sublines of Non-Small Cell Lung Cancer Cells. Int J Mol Sci 2023; 24:ijms24033042. [PMID: 36769365 PMCID: PMC9917840 DOI: 10.3390/ijms24033042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/09/2023] Open
Abstract
Radioresistance is a major obstacle for the successful therapy of many cancers, including non-small cell lung cancer (NSCLC). To elucidate the mechanism of radioresistance of NSCLC cells and to identify key molecules conferring radioresistance, the radioresistant subclones of p53 wild-type A549 and p53-deficient H1299 cell cultures were established. The transcriptional changes between parental and radioresistant NSCLC cells were investigated by RNA-seq. In total, expression levels of 36,596 genes were measured. Changes in the activation of intracellular molecular pathways of cells surviving irradiation relative to parental cells were quantified using the Oncobox bioinformatics platform. Following 30 rounds of 2 Gy irradiation, a total of 322 genes were differentially expressed between p53 wild-type radioresistant A549IR and parental A549 cells. For the p53-deficient (H1299) NSCLC cells, the parental and irradiated populations differed in the expression of 1628 genes and 1616 pathways. The expression of genes associated with radioresistance reflects the complex biological processes involved in clinical cancer cell eradication and might serve as a potential biomarker and therapeutic target for NSCLC treatment.
Collapse
|
25
|
Abozaid OAR, Rashed LA, El-Sonbaty SM, Abu-Elftouh AI, Ahmed ESA. Mesenchymal Stem Cells and Selenium Nanoparticles Synergize with Low Dose of Gamma Radiation to Suppress Mammary Gland Carcinogenesis via Regulation of Tumor Microenvironment. Biol Trace Elem Res 2023; 201:338-352. [PMID: 35138531 PMCID: PMC9823077 DOI: 10.1007/s12011-022-03146-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 02/01/2022] [Indexed: 01/11/2023]
Abstract
Breast cancer is one of the most prevalent and deadliest cancers among women in the world because of its aggressive behavior and inadequate response to conventional therapies. Mesenchymal stem cells (MSCs) combined with green nanomaterials could be an efficient tool in cell cancer therapy. This study examined the curative effects of bone marrow-derived mesenchymal stem cells (BM-MSCs) with selenium nanoparticles (SeNPs) coated with fermented soymilk and a low dose of gamma radiation (LDR) in DMBA-induced mammary gland carcinoma in female rats. DMBA-induced mammary gland carcinoma as marked by an elevation of mRNA level of cancer promoter genes (Serpin and MIF, LOX-1, and COL1A1) and serum level of VEGF, TNF-α, TGF-β, CA15-3, and caspase-3 with the reduction in mRNA level of suppressor gene (FST and ADRP). These deleterious effects were hampered after treatment with BM-MSCs (1 × 106 cells/rat) once and daily administration of SeNPs (20 mg/kg body weight) and exposure once to (0.25 Gy) LDR. Finally, MSCs, SeNPs, and LDR notably modulated the expression of multiple tumor promoters and suppressor genes playing a role in breast cancer induction and suppression.
Collapse
Affiliation(s)
- Omayma A. R. Abozaid
- Biochemistry Department, Faculty of Veterinary Medicine, Benha University, Banha, Egypt
| | - Laila A. Rashed
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Sawsan M. El-Sonbaty
- Radiation Microbiology Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | | | - Esraa S. A. Ahmed
- Radiation Biology Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Nasr City, Cairo, 11787 Egypt
| |
Collapse
|
26
|
Collagen-Specific Molecular Magnetic Resonance Imaging of Prostate Cancer. Int J Mol Sci 2022; 24:ijms24010711. [PMID: 36614152 PMCID: PMC9821004 DOI: 10.3390/ijms24010711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Constant interactions between tumor cells and the extracellular matrix (ECM) influence the progression of prostate cancer (PCa). One of the key components of the ECM are collagen fibers, since they are responsible for the tissue stiffness, growth, adhesion, proliferation, migration, invasion/metastasis, cell signaling, and immune recruitment of tumor cells. To explore this molecular marker in the content of PCa, we investigated two different tumor volumes (500 mm3 and 1000 mm3) of a xenograft mouse model of PCa with molecular magnetic resonance imaging (MRI) using a collagen-specific probe. For in vivo MRI evaluation, T1-weighted sequences before and after probe administration were analyzed. No significant signal difference between the two tumor volumes could be found. However, we detected a significant difference between the signal intensity of the peripheral tumor area and the central area of the tumor, at both 500 mm3 (p < 0.01, n = 16) and at 1000 mm3 (p < 0.01, n = 16). The results of our histologic analyses confirmed the in vivo studies: There was no significant difference in the amount of collagen between the two tumor volumes (p > 0.05), but within the tumor, higher collagen expression was observed in the peripheral area compared with the central area of the tumor. Laser ablation with inductively coupled plasma mass spectrometry further confirmed these results. The 1000 mm3 tumors contained 2.8 ± 1.0% collagen and the 500 mm3 tumors contained 3.2 ± 1.2% (n = 16). There was a strong correlation between the in vivo MRI data and the ex vivo histological data (y = −0.068x + 1.1; R2 = 0.74) (n = 16). The results of elemental analysis by inductively coupled plasma mass spectrometry supported the MRI data (y = 3.82x + 0.56; R2 = 0.79; n = 7). MRI with the collagen-specific probe in PCa enables differentiation between different tumor areas. This may help to differentiate tumor from healthy tissue, potentially identifying tumor areas with a specific tumor biology.
Collapse
|
27
|
Lu X, Xin DE, Du JK, Zou QC, Wu Q, Zhang YS, Deng W, Yue J, Fan XS, Zeng Y, Cheng X, Li X, Hou Z, Mohan M, Zhao TC, Lu X, Chang Z, Xu L, Sun Y, Zu X, Zhang Y, Chinn YE. Loss of LOXL2 Promotes Uterine Hypertrophy and Tumor Progression by Enhancing H3K36ac-Dependent Gene Expression. Cancer Res 2022; 82:4400-4413. [PMID: 36197797 DOI: 10.1158/0008-5472.can-22-0848] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/23/2022] [Accepted: 09/30/2022] [Indexed: 02/05/2023]
Abstract
UNLABELLED Lysyl oxidase-like 2 (LOXL2) is a member of the scavenger receptor cysteine-rich (SRCR) repeat carrying LOX family. Although LOXL2 is suspected to be involved in histone association and chromatin modification, the role of LOXL2 in epigenetic regulation during tumorigenesis and cancer progression remains unclear. Here, we report that nuclear LOXL2 associates with histone H3 and catalyzes H3K36ac deacetylation and deacetylimination. Both the N-terminal SRCR repeats and the C-terminal catalytic domain of LOXL2 carry redundant deacetylase catalytic activity. Overexpression of LOXL2 markedly reduced H3K36 acetylation and blocked H3K36ac-dependent transcription of genes, including c-MYC, CCND1, HIF1A, and CD44. Consequently, LOXL2 overexpression reduced cancer cell proliferation in vitro and inhibited xenograft tumor growth in vivo. In contrast, LOXL2 deficiency resulted in increased H3K36 acetylation and aberrant expression of H3K36ac-dependent genes involved in multiple oncogenic signaling pathways. Female LOXL2-deficient mice spontaneously developed uterine hypertrophy and uterine carcinoma. Moreover, silencing LOXL2 in cancer cells enhanced tumor progression and reduced the efficacy of cisplatin and anti-programmed cell death 1 (PD-1) combination therapy. Clinically, low nuclear LOXL2 expression and high H3K36ac levels corresponded to poor prognosis in uterine endometrial carcinoma patients. These results suggest that nuclear LOXL2 restricts cancer development in the female reproductive system via the regulation of H3K36ac deacetylation. SIGNIFICANCE LOXL2 loss reprograms the epigenetic landscape to promote uterine cancer initiation and progression and repress the efficacy of anti-PD-1 immunotherapy, indicating that LOXL2 is a tumor suppressor.
Collapse
Affiliation(s)
- Xufeng Lu
- Clinical Medicine Research Institute, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Zhejiang; Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Dazhuan E Xin
- Clinical Medicine Research Institute, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Zhejiang
- Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
- Institutes of Biology and Medical Sciences, Soochow University Medical College, Jiangsu, China
| | - Juanjuan K Du
- Clinical Medicine Research Institute, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Zhejiang
- Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
- Institutes of Biology and Medical Sciences, Soochow University Medical College, Jiangsu, China
| | - Quanli C Zou
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qian Wu
- Clinical Medicine Research Institute, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Zhejiang; Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Yanan S Zhang
- Clinical Medicine Research Institute, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Zhejiang; Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
- Institutes of Biology and Medical Sciences, Soochow University Medical College, Jiangsu, China
| | - Wenhai Deng
- Clinical Medicine Research Institute, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Zhejiang; Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Jicheng Yue
- Institutes of Biology and Medical Sciences, Soochow University Medical College, Jiangsu, China
| | - Xing S Fan
- Institutes of Biology and Medical Sciences, Soochow University Medical College, Jiangsu, China
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuanyuan Zeng
- Institutes of Biology and Medical Sciences, Soochow University Medical College, Jiangsu, China
| | - Xiaju Cheng
- Institutes of Biology and Medical Sciences, Soochow University Medical College, Jiangsu, China
| | - Xue Li
- Clinical Medicine Research Institute, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Zhejiang; Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Zhaoyuan Hou
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Man Mohan
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ting C Zhao
- Departments of Surgery and Medicine, Brown University School of Medicine-Rhode Island Hospital, Providence, Rhode Island
| | - Xiaomei Lu
- Cancer Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Zhijie Chang
- State Key Laboratory of Membrane Biology, Tsinghua University School of Medicine, Beijing, China
| | - Liyan Xu
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Cancer Research Center, Shantou University Medical College, Shantou, Guangdong, China
| | - Yu Sun
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiongbing Zu
- Departments of Urology and Obstetrics and Gynecology, Xiangya Hospital, Central South University, Hunan, China
| | - Yu Zhang
- Departments of Urology and Obstetrics and Gynecology, Xiangya Hospital, Central South University, Hunan, China
| | - Y Eugene Chinn
- Clinical Medicine Research Institute, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Zhejiang
- Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
- Institutes of Biology and Medical Sciences, Soochow University Medical College, Jiangsu, China
| |
Collapse
|
28
|
Anwar S, Malik JA, Ahmed S, Kameshwar VA, Alanazi J, Alamri A, Ahemad N. Can Natural Products Targeting EMT Serve as the Future Anticancer Therapeutics? MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27227668. [PMID: 36431766 PMCID: PMC9698579 DOI: 10.3390/molecules27227668] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/24/2022] [Accepted: 11/01/2022] [Indexed: 11/09/2022]
Abstract
Cancer is the leading cause of death and has remained a big challenge for the scientific community. Because of the growing concerns, new therapeutic regimens are highly demanded to decrease the global burden. Despite advancements in chemotherapy, drug resistance is still a major hurdle to successful treatment. The primary challenge should be identifying and developing appropriate therapeutics for cancer patients to improve their survival. Multiple pathways are dysregulated in cancers, including disturbance in cellular metabolism, cell cycle, apoptosis, or epigenetic alterations. Over the last two decades, natural products have been a major research interest due to their therapeutic potential in various ailments. Natural compounds seem to be an alternative option for cancer management. Natural substances derived from plants and marine sources have been shown to have anti-cancer activity in preclinical settings. They might be proved as a sword to kill cancerous cells. The present review attempted to consolidate the available information on natural compounds derived from plants and marine sources and their anti-cancer potential underlying EMT mechanisms.
Collapse
Affiliation(s)
- Sirajudheen Anwar
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Hail, Hail 81422, Saudi Arabia
- Molecular Diagnostics Unit and Personalized Treatment, University of Hail, Hail 81422, Saudi Arabia
- Correspondence:
| | - Jonaid Ahmad Malik
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Guwahati 781101, Assam, India
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar 140001, Punjab, India
| | - Sakeel Ahmed
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad 382355, Gujarat, India
| | - Verma Abhishek Kameshwar
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi 641112, Kerala, India
| | - Jowaher Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Hail, Hail 81422, Saudi Arabia
- Molecular Diagnostics Unit and Personalized Treatment, University of Hail, Hail 81422, Saudi Arabia
| | - Abdulwahab Alamri
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Hail, Hail 81422, Saudi Arabia
- Molecular Diagnostics Unit and Personalized Treatment, University of Hail, Hail 81422, Saudi Arabia
| | - Nafees Ahemad
- School of Pharmacy, Monash University Malaysia, Jalan lagoon Selatan, Bandar Sunway, Petaling Jaya 47500, Selangor DE, Malaysia
| |
Collapse
|
29
|
Li M, Zhang X, Wang M, Wang Y, Qian J, Xing X, Wang Z, You Y, Guo K, Chen J, Gao D, Zhao Y, Zhang L, Chen R, Cui J, Ren Z. Activation of Piezo1 contributes to matrix stiffness-induced angiogenesis in hepatocellular carcinoma. Cancer Commun (Lond) 2022; 42:1162-1184. [PMID: 36181398 PMCID: PMC9648387 DOI: 10.1002/cac2.12364] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 06/23/2022] [Accepted: 09/15/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Despite integrin being highlighted as a stiffness-sensor molecule in matrix stiffness-driven angiogenesis, other stiffness-sensor molecules and their mechanosensory pathways related to angiogenesis in hepatocellular carcinoma (HCC) remain obscure. Here, we explored the interplay between Piezo1 and integrin β1 in the mechanosensory pathway and their effects on HCC angiogenesis to better understand matrix stiffness-induced angiogenesis. METHODS The role of Piezo1 in matrix stiffness-induced angiogenesis was investigated using orthotopic liver cancer SD rat models with high liver stiffness background, and its clinical significance was evaluated in human HCC tissues. Matrix stiffness-mediated Piezo1 upregulation and activation were assayed using an in vitro fibronectin (FN)-coated cell culture system with different stiffness, Western blotting and Ca2+ probe. The effects of shPiezo1-conditioned medium (CM) on angiogenesis were examined by tube formation assay, wound healing assay and angiogenesis array. The underlying mechanism by which Piezo1 participated in matrix stiffness-induced angiogenesis was analyzed by microRNA quantitative real-time polymerase chain reaction (qRT-PCR), matrix stiffness measurement, dual-luciferase reporter assay, ubiquitination assay and co-immunoprecipitation. RESULTS Increased matrix stiffness significantly upregulated Piezo1 expression at both cellular and tissue levels, and high expression of Piezo1 indicated an unfavorable prognosis. High matrix stiffness also noticeably enhanced the activation level of Piezo1, similar to its expression level. Piezo1 knockdown significantly suppressed tumor growth, angiogenesis, and lung metastasis of HCC rat models with high liver stiffness background. shPiezo1-CM from HCC cells attenuated tube formation and migration abilities of vascular endothelial cells remarkably, and analysis of differentially expressed pro-angiogenic factors revealed that Piezo1 promoted the expression and secretion of vascular endothelial growth factor (VEGF), CXC chemokine ligand 16 (CXCL16) and insulin-like growth factor binding protein 2 (IGFBP2). Matrix stiffness-caused Piezo1 upregulation/activation restrained hypoxia inducible factor-1α (HIF-1α) ubiquitination, subsequently enhanced the expression of downstream pro-angiogenic factors to accelerate HCC angiogenesis. Besides, collagen 1 (COL1)-reinforced tissue stiffening resulted in more expression of Piezo1 via miR-625-5p. CONCLUSIONS This study unravels a new mechanism by which the integrin β1/Piezo1 activation/Ca2+ influx/HIF-1α ubiquitination/VEGF, CXCL16 and IGFBP2 pathway participates in matrix stiffness-driven HCC angiogenesis. Simultaneously, a positive feedback regulation loop as stiff matrix/integrin β1/miR-625-5p/Piezo1 and COL1/stiffer matrix mediates matrix stiffness-caused Piezo1 upregulation.
Collapse
Affiliation(s)
- Miao Li
- Liver Cancer InstituteZhongshan HospitalFudan University & Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghai200032P. R. China
| | - Xi Zhang
- Liver Cancer InstituteZhongshan HospitalFudan University & Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghai200032P. R. China
| | - Mimi Wang
- Liver Cancer InstituteZhongshan HospitalFudan University & Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghai200032P. R. China
| | - Yaohui Wang
- Department of RadiologyShanghai Cancer CenterFudan UniversityShanghai200032P. R. China
| | - Jiali Qian
- Department of EndocrinologyHuashan HospitalFudan UniversityShanghai200032P. R. China
| | - Xiaoxia Xing
- Liver Cancer InstituteZhongshan HospitalFudan University & Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghai200032P. R. China
| | - Zhiming Wang
- Department of OncologyZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Yang You
- Department of OncologyZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Kun Guo
- Liver Cancer InstituteZhongshan HospitalFudan University & Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghai200032P. R. China
| | - Jie Chen
- Liver Cancer InstituteZhongshan HospitalFudan University & Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghai200032P. R. China
| | - Dongmei Gao
- Liver Cancer InstituteZhongshan HospitalFudan University & Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghai200032P. R. China
| | - Yan Zhao
- Liver Cancer InstituteZhongshan HospitalFudan University & Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghai200032P. R. China
| | - Lan Zhang
- Liver Cancer InstituteZhongshan HospitalFudan University & Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghai200032P. R. China
| | - Rongxin Chen
- Liver Cancer InstituteZhongshan HospitalFudan University & Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghai200032P. R. China
| | - Jiefeng Cui
- Liver Cancer InstituteZhongshan HospitalFudan University & Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghai200032P. R. China
| | - Zhenggang Ren
- Liver Cancer InstituteZhongshan HospitalFudan University & Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghai200032P. R. China
| |
Collapse
|
30
|
The distance distribution of human microRNAs in MirGeneDB database. Sci Rep 2022; 12:17696. [PMID: 36271017 PMCID: PMC9587017 DOI: 10.1038/s41598-022-22253-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 10/12/2022] [Indexed: 01/18/2023] Open
Abstract
MicroRNAs (miRNAs) are small single-stranded non-coding RNAs around 22 nucleotide lengths found in organisms, playing an important role in cell differentiation, development, gene regulation, and apoptosis. The distance of disease miRNA biomarkers has been used to explore the association between various diseases as well as the association between virus and disease in the literature. To date, there have been no studies on deriving the distribution of the pairwise distance of human miRNAs. As the pairwise distance of miRNA biomarkers might be a useful tool in studying the disease association, in this paper, the distance distributions of human miRNAs were derived such that they could be used to measure the closeness between miRNAs. Two distance models were used to calculate the pairwise distances of 567 Homo sapiens miRNA genes accessed from the MirGeneDB database. These miRNA pairwise distances were fitted by the normal distribution, gamma distribution, empirical cumulative distribution, and the kernel density estimation method. This is the first study to provide the distance distribution of human miRNAs. The similarity of miRNA biomarkers for several diseases was examined using the derived distributions.
Collapse
|
31
|
Lysyl Oxidase Family Proteins: Prospective Therapeutic Targets in Cancer. Int J Mol Sci 2022; 23:ijms232012270. [PMID: 36293126 PMCID: PMC9602794 DOI: 10.3390/ijms232012270] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 10/03/2022] [Accepted: 10/08/2022] [Indexed: 11/06/2022] Open
Abstract
The lysyl oxidase (LOX) family, consisting of LOX and LOX-like proteins 1–4 (LOXL1–4), is responsible for the covalent crosslinking of collagen and elastin, thus maintaining the stability of the extracellular matrix (ECM) and functioning in maintaining connective tissue function, embryonic development, and wound healing. Recent studies have found the aberrant expression or activity of the LOX family occurs in various types of cancer. It has been proved that the LOX family mainly performs tumor microenvironment (TME) remodeling function and is extensively involved in tumor invasion and metastasis, immunomodulation, proliferation, apoptosis, etc. With relevant translational research in progress, the LOX family is expected to be an effective target for tumor therapy. Here, we review the research progress of the LOX family in tumor progression and therapy to provide novel insights for future exploration of relevant tumor mechanism and new therapeutic targets.
Collapse
|
32
|
Baldari S, Di Modugno F, Nisticò P, Toietta G. Strategies for Efficient Targeting of Tumor Collagen for Cancer Therapy. Cancers (Basel) 2022; 14:cancers14194706. [PMID: 36230627 PMCID: PMC9563908 DOI: 10.3390/cancers14194706] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/25/2022] Open
Abstract
Simple Summary The tumor microenvironment encompasses the cellular and extracellular matrix components that support and shape the three-dimensional framework in which solid tumors develop and grow. The extracellular matrix of the tumor is characterized by increased deposition and aberrant architecture of collagen fibers. Therefore, as a key mechanical component of the tumor microenvironment, collagen plays a critical role in cancer progression, metastasis, and therapeutic response. To boost the efficacy of current anticancer therapies, including immunotherapy, innovative approaches should take into account strategies directed against the dysregulated non-cancer cell stromal components. In the current review, we provide an overview of the principal approaches to target tumor collagen to provide therapeutic benefits. Abstract The tumor stroma, which comprises stromal cells and non-cellular elements, is a critical component of the tumor microenvironment (TME). The dynamic interactions between the tumor cells and the stroma may promote tumor progression and metastasis and dictate resistance to established cancer therapies. Therefore, novel antitumor approaches should combine anticancer and anti-stroma strategies targeting dysregulated tumor extracellular matrix (ECM). ECM remodeling is a hallmark of solid tumors, leading to extensive biochemical and biomechanical changes, affecting cell signaling and tumor tissue three-dimensional architecture. Increased deposition of fibrillar collagen is the most distinctive alteration of the tumor ECM. Consequently, several anticancer therapeutic strategies have been developed to reduce excessive tumor collagen deposition. Herein, we provide an overview of the current advances and challenges of the main approaches aiming at tumor collagen normalization, which include targeted anticancer drug delivery, promotion of degradation, modulation of structure and biosynthesis of collagen, and targeting cancer-associated fibroblasts, which are the major extracellular matrix producers.
Collapse
|
33
|
Collagen Remodeling along Cancer Progression Providing a Novel Opportunity for Cancer Diagnosis and Treatment. Int J Mol Sci 2022; 23:ijms231810509. [PMID: 36142424 PMCID: PMC9502421 DOI: 10.3390/ijms231810509] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/01/2022] [Accepted: 09/07/2022] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) is a significant factor in cancer progression. Collagens, as the main component of the ECM, are greatly remodeled alongside cancer development. More and more studies have confirmed that collagens changed from a barrier to providing assistance in cancer development. In this course, collagens cause remodeling alongside cancer progression, which in turn, promotes cancer development. The interaction between collagens and tumor cells is complex with biochemical and mechanical signals intervention through activating diverse signal pathways. As the mechanism gradually clears, it becomes a new target to find opportunities to diagnose and treat cancer. In this review, we investigated the process of collagen remodeling in cancer progression and discussed the interaction between collagens and cancer cells. Several typical effects associated with collagens were highlighted in the review, such as fibrillation in precancerous lesions, enhancing ECM stiffness, promoting angiogenesis, and guiding invasion. Then, the values of cancer diagnosis and prognosis were focused on. It is worth noting that several generated fragments in serum were reported to be able to be biomarkers for cancer diagnosis and prognosis, which is beneficial for clinic detection. At a glance, a variety of reported biomarkers were summarized. Many collagen-associated targets and drugs have been reported for cancer treatment in recent years. The new targets and related drugs were discussed in the review. The mass data were collected and classified by mechanism. Overall, the interaction of collagens and tumor cells is complicated, in which the mechanisms are not completely clear. A lot of collagen-associated biomarkers are excavated for cancer diagnosis. However, new therapeutic targets and related drugs are almost in clinical trials, with merely a few in clinical applications. So, more efforts are needed in collagens-associated studies and drug development for cancer research and treatment.
Collapse
|
34
|
Zhou YH, Guo Y, Zhu JY, Tang CY, Zhao YQ, Zhou HD. Spheroid co-culture of BMSCs with osteocytes yields ring-shaped bone-like tissue that enhances alveolar bone regeneration. Sci Rep 2022; 12:14636. [PMID: 36030312 PMCID: PMC9420131 DOI: 10.1038/s41598-022-18675-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 08/17/2022] [Indexed: 11/09/2022] Open
Abstract
Oral and maxillofacial bone defects severely impair appearance and function, and bioactive materials are urgently needed for bone regeneration. Here, we spheroid co-cultured green fluorescent protein (GFP)-labeled bone marrow stromal cells (BMSCs) and osteocyte-like MLO-Y4 cells in different ratios (3:1, 2:1, 1:1, 1:2, 1:3) or as monoculture. Bone-like tissue was formed in the 3:1, 2:1, and 1:1 co-cultures and MLO-Y4 monoculture. We found a continuous dense calcium phosphate structure and spherical calcium phosphate similar to mouse femur with the 3:1, 2:1, and 1:1 co-cultures, along with GFP-positive osteocyte-like cells encircled by an osteoid-like matrix similar to cortical bone. Flake-like calcium phosphate, which is more mature than spherical calcium phosphate, was found with the 3:1 and 2:1 co-cultures. Phosphorus and calcium signals were highest with 3:1 co-culture, and this bone-like tissue was ring-shaped. In a murine tooth extraction model, implantation of the ring-shaped bone-like tissue yielded more bone mass, osteoid and mineralized bone, and collagen versus no implantation. This tissue fabricated by spheroid co-culturing BMSCs with osteocytes yields an internal structure and mineral composition similar to mouse femur and could promote bone formation and maturation, accelerating regeneration. These findings open the way to new strategies in bone tissue engineering.
Collapse
Affiliation(s)
- Ying-Hui Zhou
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.,Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yue Guo
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.,Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Jia-Yu Zhu
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Chen-Yi Tang
- Department of Nutrition, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China
| | - Ya-Qiong Zhao
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Hou-De Zhou
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
35
|
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
|
36
|
Zhang X, Zhou W, Niu Y, Zhu S, Zhang Y, Li X, Yu C. Lysyl oxidase promotes renal fibrosis via accelerating collagen cross-link driving by β-arrestin/ERK/STAT3 pathway. FASEB J 2022; 36:e22427. [PMID: 35792886 PMCID: PMC9544652 DOI: 10.1096/fj.202200573r] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/25/2022] [Accepted: 06/10/2022] [Indexed: 11/27/2022]
Abstract
Lysyl oxidase (LOX) is a copper‐dependent monoamine oxidase whose primary function is the covalent cross‐linking of collagen in the extracellular matrix (ECM). Evidence has shown that LOX is associated with cancer and some fibrotic conditions. We recently found that serum LOX is a potential diagnostic biomarker for renal fibrosis, but the mechanism by which LOX is regulated and contributes to renal fibrosis remains unknown. The current study demonstrates the following: (1) LOX expression was increased in fibrotic kidneys including ischemia‐reperfusion injury‐(IRI‐), unilateral ureteral obstruction‐(UUO‐), and folic acid‐ (FA‐) induced fibrotic kidneys as well as in the paraffin‐embedded sections of human kidneys from the patients with renal fibrosis. (2) The increasing deposition and cross‐linking of collagen induced by LOX was observed in IRI‐, UUO‐ and FA‐kidneys. (3) LOX was regulated by the β‐arrestin‐ERK‐STAT3 pathway in renal fibrosis. STAT3 was the downstream of AT1R‐β‐arrestin‐ERK, ERK entered the nucleus and activated STAT3‐pY705 but not STAT3‐pS727. (4) STAT3 nuclear subtranslocation and binding to the LOX promoter may be responsible for the upregulation of LOX expression. (5) Pharmacologic inhibition of LOX with BAPN in vivo inhibited the upregulation of LOX, decreased collagen over cross‐linking and ameliorated renal fibrosis after ischemic injury. Collectively, these observations suggest that LOX plays an essential role in the development of renal fibrosis by catalyzing collagen over cross‐linking. Thus, strategies targeting LOX could be a new avenue in developing therapeutics against renal fibrosis.
Collapse
Affiliation(s)
- Xiaoqin Zhang
- Department of Nephrology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wenqian Zhou
- Department of Nephrology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yangyang Niu
- Department of Nephrology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Saiya Zhu
- Department of Nephrology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yingying Zhang
- Department of Nephrology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Dpartment of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Chen Yu
- Department of Nephrology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
37
|
Kim S, Lim E, Yoo K, Zhao Y, Kang J, Lim E, Shin I, Kang S, Lim HW, Lee S. Glioblastoma‐educated mesenchymal stem‐like cells promote glioblastoma infiltration via extracellular matrix remodelling in the tumour microenvironment. Clin Transl Med 2022; 12:e997. [PMID: 35908277 PMCID: PMC9339241 DOI: 10.1002/ctm2.997] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 12/01/2022] Open
Abstract
Background The biological function of mesenchymal stem‐like cells (MSLCs), a type of stromal cells, in the regulation of the tumour microenvironment is unclear. Here, we investigated the molecular mechanisms underlying extracellular matrix (ECM) remodelling and crosstalk between MSLCs and glioblastomas (GBMs) in tumour progression. Methods In vitro and in vivo co‐culture systems were used to analyze ECM remodelling and GBM infiltration. In addition, clinical databases, samples from patients with GBM and a xenografted mouse model of GBM were used. Results Previous studies have shown that the survival of patients with GBM from whom MSLCs could be isolated is substantially shorter than that of patients from whom MSLCs could not be isolated. Therefore, we determined the correlation between changes in ECM‐related gene expression in MSLC‐isolatable patients with that in MSLC non‐isolatable patients using gene set enrichment analysis (GSEA). We found that lysyl oxidase (LOX) and COL1A1 expressions increased in MSLCs via GBM‐derived clusters of differentiation 40 ligand (CD40L). Mechanistically, MSLCs are reprogrammed by the CD40L/CD40/NFκB2 signalling axis to build a tumour infiltrative microenvironment involving collagen crosslinking. Importantly, blocking of CD40L by a neutralizing antibody‐suppressed LOX expression and ECM remodelling, decreasing GBM infiltration in mouse xenograft models. Clinically, high expression of CD40L, clusters of differentiation 40 (CD40) and LOX correlated with poor survival in patients with glioma. This indicated that GBM‐educated MSLCs promote GBM infiltration via ECM remodelling in the tumour microenvironment. Conclusion Our findings provide mechanistic insights into the pro‐infiltrative tumour microenvironment produced by GBM‐educated MSLCs and highlight a potential therapeutic target that can be used for suppressing GBM infiltration.
Collapse
Affiliation(s)
- Seung‐Mo Kim
- Department of Life Science Research Institute for Natural Sciences Hanyang University Seoul Korea
| | - Eun‐Jung Lim
- Department of Life Science Research Institute for Natural Sciences Hanyang University Seoul Korea
- Memorial Sloan Kettering, Cancer Center New York New York USA
| | - Ki‐Chun Yoo
- Department of Life Science Research Institute for Natural Sciences Hanyang University Seoul Korea
- Department of Lymphoma and Myeloma Division of Cancer Medicine Center for Cancer Immunology Research The University of Texas MD Anderson Cancer Center Houston Texas USA
| | - Yi Zhao
- Department of Life Science Research Institute for Natural Sciences Hanyang University Seoul Korea
| | - Jae‐Hyeok Kang
- Department of Life Science Research Institute for Natural Sciences Hanyang University Seoul Korea
| | - Eun‐Ji Lim
- Department of Life Science Research Institute for Natural Sciences Hanyang University Seoul Korea
| | - Incheol Shin
- Department of Life Science Research Institute for Natural Sciences Hanyang University Seoul Korea
| | - Seok‐Gu Kang
- Department of Neurosurgery Brain Tumor Center, Severance Hospital Yonsei University College of Medicine Seoul Korea
| | - Han Woong Lim
- Department of Ophthalmology Hanyang University Hospital Hanyang University College of Medicine Seoul Korea
| | - Su‐Jae Lee
- Department of Life Science Research Institute for Natural Sciences Hanyang University Seoul Korea
- Fibrosis and Cancer Targeting Biotechnology FNCT Biotech Seoul Korea
| |
Collapse
|
38
|
Luthold C, Hallal T, Labbé DP, Bordeleau F. The Extracellular Matrix Stiffening: A Trigger of Prostate Cancer Progression and Castration Resistance? Cancers (Basel) 2022; 14:cancers14122887. [PMID: 35740556 PMCID: PMC9221142 DOI: 10.3390/cancers14122887] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 02/06/2023] Open
Abstract
Despite advancements made in diagnosis and treatment, prostate cancer remains the second most diagnosed cancer among men worldwide in 2020, and the first in North America and Europe. Patients with localized disease usually respond well to first-line treatments, however, up to 30% develop castration-resistant prostate cancer (CRPC), which is often metastatic, making this stage of the disease incurable and ultimately fatal. Over the last years, interest has grown into the extracellular matrix (ECM) stiffening as an important mediator of diseases, including cancers. While this process is increasingly well-characterized in breast cancer, a similar in-depth look at ECM stiffening remains lacking for prostate cancer. In this review, we scrutinize the current state of literature regarding ECM stiffening in prostate cancer and its potential association with disease progression and castration resistance.
Collapse
Affiliation(s)
- Carole Luthold
- Centre de Recherche sur le Cancer, Université Laval, Québec, QC G1R 3S3, Canada;
- Division of Oncology, Centre de Recherche du CHU de Québec-Université Laval, Hôtel-Dieu de Québec, Québec, QC G1R 3S3, Canada
| | - Tarek Hallal
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada;
| | - David P. Labbé
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada;
- Division of Urology, Department of Surgery, McGill University, Montréal, QC H4A 3J1, Canada
- Correspondence: (D.P.L.); (F.B.)
| | - François Bordeleau
- Centre de Recherche sur le Cancer, Université Laval, Québec, QC G1R 3S3, Canada;
- Division of Oncology, Centre de Recherche du CHU de Québec-Université Laval, Hôtel-Dieu de Québec, Québec, QC G1R 3S3, Canada
- Département de Biologie Moléculaire, Biochimie Médicale et Pathologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Correspondence: (D.P.L.); (F.B.)
| |
Collapse
|
39
|
Shukla P, Yeleswarapu S, Heinrich M, Prakash J, Pati F. Mimicking Tumor Microenvironment by 3D Bioprinting: 3D Cancer Modeling. Biofabrication 2022; 14. [PMID: 35512666 DOI: 10.1088/1758-5090/ac6d11] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 05/05/2022] [Indexed: 11/12/2022]
Abstract
The tumor microenvironment typically comprises cancer cells, tumor vasculature, stromal components like fibroblasts, and host immune cells that assemble to support tumorigenesis. However, preexisting classic cancer models like 2D cell culture methods, 3D cancer spheroids, and tumor organoids seem to lack essential tumor microenvironment components. 3D bioprinting offers enormous advantages for developing in vitro tumor models by allowing user-controlled deposition of multiple biomaterials, cells, and biomolecules in a predefined architecture. This review highlights the recent developments in 3D cancer modeling using different bioprinting techniques to recreate the TME. 3D bioprinters enable fabrication of high-resolution microstructures to reproduce TME intricacies. Furthermore, 3D bioprinted models can be applied as a preclinical model for versatile research applications in the tumor biology and pharmaceutical industries. These models provide an opportunity to develop high-throughput drug screening platforms and can further be developed to suit individual patient requirements hence giving a boost to the field of personalized anti-cancer therapeutics. We underlined the various ways the existing studies have tried to mimic the TME, mimic the hallmark events of cancer growth and metastasis within the 3D bioprinted models and showcase the 3D drug-tumor interaction and further utilization of such models to develop personalized medicine.
Collapse
Affiliation(s)
- Priyanshu Shukla
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Hyderabad, Telangana, 502285, INDIA
| | - Sriya Yeleswarapu
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Hyderabad, Telangana, 502285, INDIA
| | - Marcel Heinrich
- Department of Biomaterials, Science and Technology, University of Twente Faculty of Science and Technology, Department of Biomaterials, Science and Technology, Faculty of Science and Technology, University of Twente, Drienerlolaan 5, 7500AE, Enschede, The Netherlands, Enschede, Overijssel, 7500 AE, NETHERLANDS
| | - Jai Prakash
- Department of Biomaterials, Science and Technology, University of Twente Faculty of Science and Technology, Department of Biomaterials, Science and Technology, Faculty of Science and Technology, University of Twente, Drienerlolaan 5, 7500AE, Enschede, The Netherlands, Enschede, Overijssel, 7500 AE, NETHERLANDS
| | - Falguni Pati
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Hyderabad, Telangana, 502285, INDIA
| |
Collapse
|
40
|
Ren P, Niu X, Zhao R, Liu J, Ren W, Dai H, Chen J, Yan J, Li B, Shao Y, Bai Y, Han P. Long non-coding RNA AGAP2-AS1 promotes cell proliferation and invasion through regulating miR-193a-3p/LOXL4 axis in laryngeal squamous cell carcinoma. Cell Cycle 2022; 21:697-707. [PMID: 35113007 PMCID: PMC8973330 DOI: 10.1080/15384101.2021.2016197] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is an aggressive malignancy with highly mortality rate. Long non-coding RNA (lncRNA) AGAP2-AS1 is an identified oncogene in several types of cancers. However, the role of AGAP2-AS1 in LSCC remains unclear. The expression levels of AGAP2-AS1 in LSCC tissues and cell lines were measured using qRT-PCR. AGAP2-AS1 was knocked down in LSCC cells through transfection with siRNA-AGAP2-AS1. Cell proliferation and invasion were detected using MTT and transwell assays. Dual-luciferase reporter gene assay was performed to confirm the interaction with AGAP2-AS1 and downstream genes. Our results showed that AGAP2-AS1 expression was remarkably increased in human LSCC tissues and cell lines. Knockdown of AGAP2-AS1 significantly inhibited the proliferation and invasion of LSCC cells. In addition, AGAP2-AS1 sponged miR-193a-3p and regulated its expression in LSCC cells. Inhibition of miR-193a-3p reversed the effects of AGAP2-AS1 knockdown on LSCC cells. Furthermore, Lysyl oxidase-like 4 (LOXL4) was a target gene of miR-193a-3p and the role of miR-193a-3p was mediated by LOXL4. In conclusion, these findings suggest that knockdown of AGAP2-AS1 inhibited the proliferation and invasion of LSCC cells through regulating the miR-193a-3p/LOXL4 axis.
Collapse
Affiliation(s)
- Pengyu Ren
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China,Department of Neurosurgery, Second Affiliated Hospital of Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China
| | - Xiaorong Niu
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China
| | - Ruimin Zhao
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China
| | - Junsong Liu
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China
| | - Wanli Ren
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China
| | - Hao Dai
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China
| | - Jiayu Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China
| | - Jinfeng Yan
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China
| | - Baiya Li
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China
| | - Yuan Shao
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China
| | - Yanxia Bai
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China,CONTACT Yanxia Bai
| | - Peng Han
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi, China,Peng Han Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi710061, China
| |
Collapse
|
41
|
Jiang C, Wang M, Yao W, Lv G, Liu X, Wang G. Comprehensive Analysis on Prognosis and Immune Infiltration of Lysyl Oxidase Family Members in Pancreatic Adenocarcinoma With Experimental Verification. Front Mol Biosci 2022; 9:778857. [PMID: 35433829 PMCID: PMC9010946 DOI: 10.3389/fmolb.2022.778857] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Pancreatic adenocarcinoma (PDAC) is the most aggressive among all solid malignancies with delayed disease detection and limited effective treatment. However, due to the intricate heterogeneity and exclusive tumor microenvironment (TME), the development of effective therapy has been facing enormous challenges. The lysyl oxidases (LOXs) underpin the shaping of the TME to promote cancer growth, metastasis and modulate response to treatment.Materials and Methods: The mRNA expression, prognostic, and clinicopathological data for LOXs in PDAC from multiple open-access databases were summarized and analyzed. The protein expression was verified by immunohistochemistry (IHC). Co-expressed genes of LOXs were predicted and elaborated by LinkedOmics. Functional enrichment analysis of LOXs co-expressed genes was performed using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). TIMER and TISIDB were applied to analyze the relationship between LOXs expression and immune infiltration.Results: The mRNA expression levels of LOX, LOXL1 and LOXL2 were significantly higher in PDAC, the expression levels of LOXL3 and LOXL4 were contrary in different databases. High mRNA levels of LOX and LOXL2 were associated with advanced PDAC stage, while elevated LOX and LOXL3 expression correlated with high tumor grade. The IHC staining showed higher expression levels of LOX, LOXL1 and LOXL2, lower expression level of LOXL3 in PDAC tissues, while the protein expression of LOXL4 made no difference. Functional enrichment analysis showed a close relationship with extracellular matrix (ECM) regulation, except that LOXL3 and its ligands were highly associated with immune-related functions. Further analysis suggested that LOX and LOXL3 strongly correlated with tumor-infiltrating lymphocytes (TILs), various immune signatures, and immune checkpoints. Finally, survival analysis revealed high LOX and LOXL2 expression predicted worse overall survival (OS), progression-free interval (PFI), and disease-specific survival (DSS).Conclusion: These findings indicated that the LOX family, especially LOX and LOXL2, might have a prospective value in PDAC oncogenesis, and they may become prognostic biomarkers, revealing a promising field in targeted therapy.
Collapse
Affiliation(s)
- Chao Jiang
- Department of Hepatobiliary Pancreatic Surgery I, The First Hospital of Jilin University, Changchun, China
- Multi-Organ Transplant Program, University Health Network, Toronto, ON, Canada
| | - Meng Wang
- Department of Hepatobiliary Pancreatic Surgery I, The First Hospital of Jilin University, Changchun, China
| | - Weikai Yao
- Department of Pathology, The First Hospital of Jilin University, Changchun, China
| | - Guoyue Lv
- Department of Hepatobiliary Pancreatic Surgery I, The First Hospital of Jilin University, Changchun, China
| | - Xueyan Liu
- Cardiovascular Department, China-Japan Union Hospital of Jilin University, Changchun, China
- *Correspondence: Xueyan Liu, ; Guangyi Wang,
| | - Guangyi Wang
- Department of Hepatobiliary Pancreatic Surgery I, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Xueyan Liu, ; Guangyi Wang,
| |
Collapse
|
42
|
Natural Compounds Targeting Cancer-Associated Fibroblasts against Digestive System Tumor Progression: Therapeutic Insights. Biomedicines 2022; 10:biomedicines10030713. [PMID: 35327514 PMCID: PMC8945097 DOI: 10.3390/biomedicines10030713] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 01/27/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) are critical for cancer occurrence and progression in the tumor microenvironment (TME), due to their versatile roles in extracellular matrix remodeling, tumor–stroma crosstalk, immunomodulation, and angiogenesis. CAFs are the most abundant stromal component in the TME and undergo epigenetic modification and abnormal signaling cascade activation, such as transforming growth factor-β (TGF-β) and Wnt pathways that maintain the distinct phenotype of CAFs, which differs from normal fibroblasts. CAFs have been considered therapeutic targets due to their putative oncogenic functions. Current digestive system cancer treatment strategies often result in lower survival outcomes and fail to prevent cancer progression; therefore, comprehensive characterization of the tumor-promoting and -restraining CAF activities might facilitate the design of new therapeutic approaches. In this review, we summarize the enormous literature on natural compounds that mediate the crosstalk of CAFs with digestive system cancer cells, discuss how the biology and the multifaceted functions of CAFs contribute to cancer progression, and finally, pave the way for CAF-related antitumor therapies.
Collapse
|
43
|
Shi R, Zhang Z, Zhu A, Xiong X, Zhang J, Xu J, Sy MS, Li C. Targeting Type I Collagen for Cancer Treatment. Int J Cancer 2022; 151:665-683. [PMID: 35225360 DOI: 10.1002/ijc.33985] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 11/07/2022]
Abstract
Collagen is the most abundant protein in animals. Interactions between tumor cells and collagen influence every step of tumor development. Type I collagen is the main fibrillar collagen in the extracellular matrix and is frequently up-regulated during tumorigenesis. The binding of type I collagen to its receptors on tumor cells promotes tumor cell proliferation, epithelial-mesenchymal transition, and metastasis. Type I collagen also regulates the efficacy of tumor therapies, such as chemotherapy, radiotherapy, and immunotherapy. Furthermore, type I collagen fragments are diagnostic markers of metastatic tumors and have prognostic value. Inhibition of type I collagen synthesis has been reported to have anti-tumor effects in animal models. However, collagen has also been shown to possess anti-tumor activity. Therefore, the roles that type I collagen plays in tumor biology are complex and tumor type-dependent. In this review, we discuss the expression and regulation of synthesis of type I collagen, as well as the role up-regulated type I collagen plays in various stages of cancer progression. We also discuss the role of collagen in tumor therapy. Finally, we highlight several recent approaches targeting type I collagen for cancer treatment. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Run Shi
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, Guangzhou, China
| | - Zhe Zhang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, Guangzhou, China
| | - Ankai Zhu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, Guangzhou, China
| | - Xingxing Xiong
- Department of Operating Room, Jiangxi Cancer Hospital of Nanchang University, Nanchang, China
| | - Jie Zhang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, Guangzhou, China
| | - Jiang Xu
- Department of Stomatology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Man-Sun Sy
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Chaoyang Li
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, Guangzhou, China
| |
Collapse
|
44
|
Mohamadzaheri M, Cheraghi H, Shirani D, Hatamkhani A. Relationship between plasma cell-free DNA changes and lysyl oxidase during the treatment and prognosis of canine transmissible venereal tumors. BMC Vet Res 2022; 18:76. [PMID: 35189882 PMCID: PMC8862336 DOI: 10.1186/s12917-022-03173-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 02/09/2022] [Indexed: 11/22/2022] Open
Abstract
Background Transmissible venereal tumors (TVT) are a wide range of canine tumors for which there are no effective markers to monitor the therapeutic response in real-time. Circulating biomarkers can be valuable in early cancer diagnosis and prognosis. Accordingly, this study aimed to investigate the significance of the cell-free DNA (cfDNA) and cfDNA integrity index to monitor the response of TVTs to vincristine and compare them with lysyl oxidase activity. Plasma and sera were collected from fifteen male dogs within four weeks before drug administration. The analytical method was mainly based on the quantitative polymerase chain reaction (qPCR) technique for short and long cfDNAs and lysyl oxidase activity was measured in serum. Results The results of the cfDNA integrity index showed a significant (p < 0.05) difference in the baseline concentration compared to the second and third weeks (with cut-off values of 1.118 and 93.33% specificity). The cfDNA integrity index increased over time due to the reduction of short cfDNAs in the first week after treatment. Lysyl oxidase activity increased during the fourth week (p < 0.001), but there were no significant differences in the other weeks compared to the baseline. The ROC analysis of lysyl oxidase revealed high sensitivity (100%) and specificity (90%) on the second and third weeks compared to the baseline. Multivariate analysis between cfDNA integrity index and lysyl oxidase showed significant correlation (p < 0.05) only in baseline results. Conclusions Overall, short cfDNA, the cfDNA integrity index, and lysyl oxidase activity can be proposed as diagnostic biomarkers and putative prognostic candidates in TVT patients. These biomarkers can be combined with cytology to quickly diagnose TVT. Supplementary Information The online version contains supplementary material available at 10.1186/s12917-022-03173-z.
Collapse
Affiliation(s)
- Mona Mohamadzaheri
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Razi University, Kermanshah, Iran
| | - Hadi Cheraghi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Razi University, Kermanshah, Iran.
| | - Darioush Shirani
- Department of Small Animal Internal Medicine, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | |
Collapse
|
45
|
Meng K, Hu X, Zheng G, Qian C, Xin Y, Guo H, He R, Ge M, Xu J. Identification of prognostic biomarkers for papillary thyroid carcinoma by a weighted gene co‐expression network analysis. Cancer Med 2022; 11:2006-2019. [PMID: 35152572 PMCID: PMC9089218 DOI: 10.1002/cam4.4602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/13/2022] Open
Abstract
Aim Methods Results Conclusions
Collapse
Affiliation(s)
- Kexin Meng
- Otolaryngology& Head and Neck Center, Cancer Center, Department of Head and Neck Surgery Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College) Hangzhou Zhejiang China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province Hangzhou Zhejiang China
| | - Xiaotian Hu
- Otolaryngology& Head and Neck Center, Cancer Center, Department of Head and Neck Surgery Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College) Hangzhou Zhejiang China
- Qingdao University Qingdao Shandong China
| | - Guowan Zheng
- Otolaryngology& Head and Neck Center, Cancer Center, Department of Head and Neck Surgery Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College) Hangzhou Zhejiang China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province Hangzhou Zhejiang China
| | - Chenhong Qian
- Otolaryngology& Head and Neck Center, Cancer Center, Department of Head and Neck Surgery Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College) Hangzhou Zhejiang China
- Bengbu Medical College Bengbu China
| | - Ying Xin
- Otolaryngology& Head and Neck Center, Cancer Center, Department of Head and Neck Surgery Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College) Hangzhou Zhejiang China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province Hangzhou Zhejiang China
| | - Haiwei Guo
- Otolaryngology& Head and Neck Center, Cancer Center, Department of Head and Neck Surgery Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College) Hangzhou Zhejiang China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province Hangzhou Zhejiang China
| | - Ru He
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College Hangzhou China
| | - Minghua Ge
- Otolaryngology& Head and Neck Center, Cancer Center, Department of Head and Neck Surgery Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College) Hangzhou Zhejiang China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province Hangzhou Zhejiang China
| | - Jiajie Xu
- Otolaryngology& Head and Neck Center, Cancer Center, Department of Head and Neck Surgery Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College) Hangzhou Zhejiang China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province Hangzhou Zhejiang China
| |
Collapse
|
46
|
MiR-29a Curbs Hepatocellular Carcinoma Incidence via Targeting of HIF-1α and ANGPT2. Int J Mol Sci 2022; 23:ijms23031636. [PMID: 35163556 PMCID: PMC8835722 DOI: 10.3390/ijms23031636] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 12/11/2022] Open
Abstract
A high-fat diet is responsible for hepatic fat accumulation that sustains chronic liver damage and increases the risks of steatosis and hepatocellular carcinoma (HCC). MicroRNA-29a (miR-29a), a key regulator of cellular behaviors, is present in anti-fibrosis and modulator tumorigenesis. However, the increased transparency of the correlation between miR-29a and the progression of human HCC is still further investigated. In this study, we predicted HIF-1α and ANGPT2 as regulators of HCC by the OncoMir cancer database and showed a strong positive correlation with HIF-1α and ANGPT2 gene expression in HCC patients. Mice fed the western diet (WD) while administered CCl4 for 25 weeks induced chronic liver damage and higher HCC incidence than without fed WD mice. HCC section staining revealed signaling upregulation in ki67, severe fibrosis, and steatosis in WD and CCl4 mice and detected Col3a1 gene expressions. HCC tissues significantly attenuated miR-29a but increased in HIF-1α, ANGPT2, Lox, Loxl2, and VEGFA expression. Luciferase activity analysis confirms that miR-29a specific binding 3′UTR of HIF-1α and ANGPT2 to repress expression. In summary, miR-29a control HIF-1α and ANGPT2 signaling in HCC formation. This study insight into a novel molecular pathway by which miR-29a targeting HIF-1α and ANGPT2 counteracts the incidence of HCC development.
Collapse
|
47
|
Wang L, Cao S, Zhai R, Zhao Y, Song G. Systematic Analysis of Expression and Prognostic Values of Lysyl Oxidase Family in Gastric Cancer. Front Genet 2022; 12:760534. [PMID: 35126449 PMCID: PMC8812723 DOI: 10.3389/fgene.2021.760534] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 12/16/2021] [Indexed: 01/14/2023] Open
Abstract
Background: Gastric cancer (GC) remains the fifth most commonly diagnosed malignancy worldwide, with a poor prognosis. The lysyl oxidase (LOX) family, a type of secreted copper-dependent amine oxidases, is comprised of LOX and four LOX-like (LOXL) 1–4 isoforms and has been reported to be dysregulated in a number of different type cancers. However, the diverse expression patterns and prognostic values of LOX family in GC have yet to be systematically analyzed. Methods: ONCOMINE, GEPIA, UALCAN, Kaplan–Meier Plotter, LOGpc, cBioPortal, GeneMANIA and Metascape databases were utilized in this study to analyze the expression, prognostic values, mutations and functional networks of LOX family in GC. Results: The mRNA expression levels of LOX, LOXL1 and LOXL2 were significantly higher in GC, the expression level of LOXL3 was contrary in different databases, while the expression level of LOXL4 made no difference; the expression levels of LOX, LOXL1 and LOXL3 were higher in stages 2–4 than that of normal tissues and stage 1, while the mRNA level of LOXL2 in stage 1–4 was higher than normal tissues; patients with high expression of LOX and LOXL 2-4 had poor OS; the genes correlated with LOX and LOXL2 were enriched in extracellular matrix organization, vasculature development and skeletal system development. Conclusion: Our results indicated that the LOX family, especially LOX and LOXL2, might play an important role in GC oncogenesis, and they may become biomarkers for predicting tumor prognosis and potential targets for tumor therapy.
Collapse
Affiliation(s)
- Li Wang
- Department of Gastrointestinal Surgery, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Shan Cao
- Department of Respiratory, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Rujun Zhai
- Department of Gastrointestinal Surgery, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yang Zhao
- Radiology Department, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Guodong Song
- Department of Gastrointestinal Surgery, The Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
48
|
Lysyl-Oxidase Dependent Extracellular Matrix Stiffness in Hodgkin Lymphomas: Mechanical and Topographical Evidence. Cancers (Basel) 2022; 14:cancers14010259. [PMID: 35008423 PMCID: PMC8750937 DOI: 10.3390/cancers14010259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/30/2021] [Accepted: 01/03/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Alterations of the composition and architecture of the extracellular matrix (ECM), leading to increased stiffness, is known to condition development, invasiveness and severity of neoplasms. In this study, we report increased lymph node (LN) stiffness in human lymphomas, measured by LN elastometry or by computerized imaging of bioptic specimens. Stiffness matched to lymphoma histotype and grading. The enzyme lysyl oxidase (LOX) is involved in the rise of collagen cross-linking in Hodgkin lymphomas, while altered architecture, shown by scanning electron microscopy and polarized light microscopy is involved in advanced follicular lymphomas. Based on these data, digital pathology may help in the staging of lymphomas, and lysyl oxidase may represent a target for therapy in Hodgkin lymphomas. Abstract Purpose: The biochemical composition and architecture of the extracellular matrix (ECM) is known to condition development and invasiveness of neoplasms. To clarify this point, we analyzed ECM stiffness, collagen cross-linking and anisotropy in lymph nodes (LN) of Hodgkin lymphomas (HL), follicular lymphomas (FL) and diffuse large B-cell lymphomas (DLBCL), compared with non-neoplastic LN (LDN). Methods and Results: We found increased elastic (Young’s) modulus in HL and advanced FL (grade 3A) over LDN, FL grade 1–2 and DLBCL. Digital imaging evidenced larger stromal areas in HL, where increased collagen cross-linking was found; in turn, architectural modifications were documented in FL3A by scanning electron microscopy and enhanced anisotropy by polarized light microscopy. Interestingly, HL expressed high levels of lysyl oxidase (LOX), an enzyme responsible for collagen cross-linking. Using gelatin scaffolds fabricated with a low elastic modulus, comparable to that of non-neoplastic tissues, we demonstrated that HL LN-derived mesenchymal stromal cells and HL cells increased the Young’s modulus of the extracellular microenvironment through the expression of LOX. Indeed, LOX inhibition by β-aminopropionitrile prevented the gelatin stiffness increase. Conclusions: These data indicate that different mechanical, topographical and/or architectural modifications of ECM are detectable in human lymphomas and are related to their histotype and grading.
Collapse
|
49
|
Pielhop TP, Popp C, Knierim D, Margaria P, Maiß E. Three new mycoviruses identified in the apple replant disease (ARD)-associated fungus Rugonectria rugulosa. Virus Genes 2022; 58:423-435. [PMID: 35841525 PMCID: PMC9477930 DOI: 10.1007/s11262-022-01924-6] [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: 02/13/2022] [Accepted: 06/25/2022] [Indexed: 02/05/2023]
Abstract
In this study, three new mycoviruses were identified co-infecting the apple replant disease (ARD)-associated root endophyte Rugonectria rugulosa. After dsRNA extraction, six viral fragments were visualized. Four fragments belong to a quadrivirus, which has a genome size of 17,166 bp. Each of the fragments of this quadrivirus has a single ORF encoding a protein. Two of these proteins are coat protein subunits, one ORF encodes the RdRp, and one protein has an unknown function. This virus was tentatively named rugonectria rugulosa quadrivirus 1 (RrQV1) as a member of the proposed new species Quadrivirus rugonectria. Another fragment represents the dsRNA intermediate form of a + ssRNA mitovirus with a genome size of 2410 nt. This virus encodes an RdRp and is tentatively called rugonectria rugulosa mitovirus 1 (RrMV1). RrMV1 is suggested as a member of a new species with the proposed name Mitovirus rugonectria. The sixth fragment belongs to the genome of an unclassified dsRNA virus tentatively called rugonectria rugulosa dsRNA virus 1 (RrV1). The monopartite dsRNA genome of RrV1 has a length of 8964 bp and contains two ORFs encoding a structure/gag protein and an RdRp. Full genomic sequences were determined and the genome structure as well as molecular properties are presented. After phylogenetic studies and sequence identity analyses, all three isolates are proposed as new mycoviruses. The results help to improve the understanding of the complexity of the factors involved in ARD and support the interest in mycoviral research. Subsequent analyses need to focus on the impact of mycoviruses on the biology and pathogenicity of ARD-associated fungi. The results of such studies could contribute to the development of mitigation strategies against the disease.
Collapse
Affiliation(s)
- Tom P. Pielhop
- Institute of Horticultural Production Systems, Department of Phytomedicine, Leibniz University Hannover, Herrenhäuser Str. 2, 30419 Hannover, Germany
| | - Carolin Popp
- Institute of Horticultural Production Systems, Department of Phytomedicine, Leibniz University Hannover, Herrenhäuser Str. 2, 30419 Hannover, Germany
| | - Dennis Knierim
- Leibniz Institute DSMZ, German Collection of Microorganisms and Cell Cultures, Inhoffenstraße 7 B, 38124 Brunswick, Germany
| | - Paolo Margaria
- Leibniz Institute DSMZ, German Collection of Microorganisms and Cell Cultures, Inhoffenstraße 7 B, 38124 Brunswick, Germany
| | - Edgar Maiß
- Institute of Horticultural Production Systems, Department of Phytomedicine, Leibniz University Hannover, Herrenhäuser Str. 2, 30419 Hannover, Germany
| |
Collapse
|
50
|
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
|