1
|
Ottaviano KE, Subbaram S, Wu L, Stahl K, Mastrangelo AJ, Lee H, DiPersio CM. Integrin α3β1 Is Not Required for Onset of Dysplasia in Genetic Model of Colon Cancer but Promotes Motility of Colon Cancer Cells. Cancers (Basel) 2025; 17:371. [PMID: 39941740 PMCID: PMC11815772 DOI: 10.3390/cancers17030371] [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: 12/08/2024] [Revised: 01/13/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND/OBJECTIVES The progression of colorectal cancer through clinically and histopathologically well-defined stages is driven by specific mutations that activate oncogenes or inactivate tumor-suppressor genes. In addition, pre-cancerous/cancer cells respond to cues from the tissue microenvironment that support tumorigenesis and progression, many of which are transmitted through integrin receptors for the extracellular matrix. Integrin α3β1 has pro-tumorigenic/pro-metastatic roles in many cancers, but it also has suppressive roles in some cancers or at specific stages of progression, indicating that its potential value as a therapeutic target cannot be extrapolated across cancer types or stages. In this study, we investigated roles for α3β1 in colorectal cancer using cellular and genetic models that represent different stages. METHODS We generated mice with colon-specific α3 knockout in a tamoxifen-inducible model of KRAS-mutated colorectal cancer to assess the effects of α3β1 ablation on early dysplasia. We also used siRNA to suppress α3β1 in human colorectal cancer cells, then assessed effects on motility and invasion in vitro. RESULTS Genetic deletion of α3β1 in the colon did not alter dysplasia in mice predisposed to KRAS-mutated colorectal cancer, and it was accompanied by an increase in the colocalization of α6 integrin with laminin-332 (a matrix ligand for both integrins), suggesting functional compensation. However, suppression of α3β1 caused an approximately 40% to 60% reduction in the motility/invasion of human colorectal cancer cells. CONCLUSIONS Our findings that α3β1 is not required for pre-cancerous dysplasia but promotes colorectal cancer cell motility/invasion indicate an important role for pro-migratory functions of this integrin at later stages of progression when cells invade from the primary tumor, suggesting that strategies to target α3β1 in colorectal cancer should be aimed at distinct stages of disease progression.
Collapse
Affiliation(s)
- Kathryn E. Ottaviano
- Department of Surgery, Albany Medical College, Albany, NY 12208, USA; (K.E.O.); (S.S.); (L.W.)
| | - Sita Subbaram
- Department of Surgery, Albany Medical College, Albany, NY 12208, USA; (K.E.O.); (S.S.); (L.W.)
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (K.S.); (A.J.M.)
- Department of Biology, Union College, Schenectady, NY 12308, USA
| | - Lei Wu
- Department of Surgery, Albany Medical College, Albany, NY 12208, USA; (K.E.O.); (S.S.); (L.W.)
| | - Kiley Stahl
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (K.S.); (A.J.M.)
| | - Antoinette J. Mastrangelo
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (K.S.); (A.J.M.)
| | - Hwajeong Lee
- Department of Pathology, Albany Medical College, Albany, NY 12208, USA;
| | - C. Michael DiPersio
- Department of Surgery, Albany Medical College, Albany, NY 12208, USA; (K.E.O.); (S.S.); (L.W.)
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (K.S.); (A.J.M.)
| |
Collapse
|
2
|
WalyEldeen AA, Sabet S, Anis SE, Stein T, Ibrahim AM. FBLN2 is associated with basal cell markers Krt14 and ITGB1 in mouse mammary epithelial cells and has a preferential expression in molecular subtypes of human breast cancer. Breast Cancer Res Treat 2024; 208:673-686. [PMID: 39110274 PMCID: PMC11522194 DOI: 10.1007/s10549-024-07447-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Fibulin-2 (FBLN2) is a secreted extracellular matrix (ECM) glycoprotein and has been identified in the mouse mammary gland, in cap cells of terminal end buds (TEBs) during puberty, and around myoepithelial cells during early pregnancy. It is required for basement membrane (BM) integrity in mammary epithelium, and its loss has been associated with human breast cancer invasion. Herein, we attempted to confirm the relevance of FBLN2 to myoepithelial phenotype in mammary epithelium and to assess its expression in molecular subtypes of human breast cancer. METHODS The relationship between FBLN2 expression and epithelial markers was investigated in pubertal mouse mammary glands and the EpH4 mouse mammary epithelial cell line using immunohistochemistry, immunocytochemistry, and immunoblotting. Human breast cancer mRNA data from the METABRIC and TCGA datasets from Bioportal were analyzed to assess the association of Fbln2 expression with epithelial markers, and with molecular subtypes. Survival curves were generated using data from the METABRIC dataset and the KM databases. RESULTS FBLN2 knockdown in mouse mammary epithelial cells was associated with a reduction in KRT14 and an increase in KRT18. Further, TGFβ3 treatment resulted in the upregulation of FBLN2 in vitro. Meta-analyses of human breast cancer datasets from Bioportal showed a higher expression of Fbln2 mRNA in claudin-low, LumA, and normal-like breast cancers compared to LumB, Her2 +, and Basal-like subgroups. Fbln2 mRNA levels were positively associated with mesenchymal markers, myoepithelial markers, and markers of epithelial-mesenchymal transition. Higher expression of Fbln2 mRNA was associated with better prognosis in less advanced breast cancer and this pattern was reversed in more advanced lesions. CONCLUSION With further validation, these observations may offer a molecular prognostic tool for human breast cancer for more personalized therapeutic approaches.
Collapse
Affiliation(s)
| | - Salwa Sabet
- Department of Zoology, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Shady E Anis
- Department of Pathology, Faculty of Medicine, Cairo University, Cairo, 11562, Egypt
| | - Torsten Stein
- Institute of Veterinary Biochemistry, Freie Universität Berlin, 14163, Berlin, Germany
- Institute of Cancer Sciences, College of MVLS, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Ayman M Ibrahim
- Department of Zoology, Faculty of Science, Cairo University, Giza, 12613, Egypt.
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Aswan, Egypt.
| |
Collapse
|
3
|
Longmate WM. The epidermal integrin-mediated secretome regulates the skin microenvironment during tumorigenesis and repair. Matrix Biol 2024; 134:175-183. [PMID: 39491760 PMCID: PMC11585437 DOI: 10.1016/j.matbio.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/16/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
Integrins are cellular transmembrane receptors that physically connect the cytoskeleton with the extracellular matrix. As such, they are positioned to mediate cellular responses to microenvironmental cues. Importantly, integrins also regulate their own microenvironment through secreted factors, also known as the integrin-mediated secretome. Epidermal integrins, or integrins expressed by keratinocytes of the skin epidermis, regulate the cutaneous microenvironment through the contribution of matrix components, via proteolytic matrix remodeling, or by mediating factors like cytokines and growth factors that can promote support for nearby but distinct cells of the stroma, such as immune cells, endothelial cells, and fibroblasts. This role for integrins is enhanced during both pathological and repair tissue remodeling processes, such as tumor growth and progression and wound healing. This review will discuss examples of how the epithelial integrin-mediated secretome can regulate the tissue microenvironment. Although different epithelial integrins in various contexts will be explored, emphasis will be given to epidermal integrins that regulate the secretome during wound healing and cutaneous tumor progression. Epidermal integrin α3β1 is of particular focus as well, since this integrin has been revealed as a key regulator of the keratinocyte secretome.
Collapse
Affiliation(s)
- Whitney M Longmate
- Department of Surgery, Albany Medical College, Albany, NY 12208, USA; Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.
| |
Collapse
|
4
|
Miskin RP, DiPersio CM. Roles for epithelial integrin α3β1 in regulation of the microenvironment during normal and pathological tissue remodeling. Am J Physiol Cell Physiol 2024; 326:C1308-C1319. [PMID: 38497112 PMCID: PMC11371326 DOI: 10.1152/ajpcell.00128.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/08/2024] [Accepted: 03/08/2024] [Indexed: 03/19/2024]
Abstract
Integrin receptors for the extracellular matrix activate intracellular signaling pathways that are critical for tissue development, homeostasis, and regeneration/repair, and their loss or dysregulation contributes to many developmental defects and tissue pathologies. This review will focus on tissue remodeling roles for integrin α3β1, a receptor for laminins found in the basement membranes (BMs) that underlie epithelial cell layers. As a paradigm, we will discuss literature that supports a role for α3β1 in promoting ability of epidermal keratinocytes to modify their tissue microenvironment during skin development, wound healing, or tumorigenesis. Preclinical and clinical studies have shown that this role depends largely on ability of α3β1 to govern the keratinocyte's repertoire of secreted proteins, or the "secretome," including 1) matrix proteins and proteases involved in matrix remodeling and 2) paracrine-acting growth factors/cytokines that stimulate other cells with important tissue remodeling functions (e.g., endothelial cells, fibroblasts, inflammatory cells). Moreover, α3β1 signaling controls gene expression that helps epithelial cells carry out these functions, including genes that encode secreted matrix proteins, proteases, growth factors, or cytokines. We will review what is known about α3β1-dependent gene regulation through both transcription and posttranscriptional mRNA stability. Regarding the latter, we will discuss examples of α3β1-dependent alternative splicing (AS) or alternative polyadenylation (APA) that prevents inclusion of cis-acting mRNA sequences that would otherwise target the transcript for degradation via nonsense-mediated decay or destabilizing AU-rich elements (AREs) in the 3'-untranslated region (3'-UTR). Finally, we will discuss prospects and anticipated challenges of exploiting α3β1 as a clinical target for the treatment of cancer or wound healing.
Collapse
Affiliation(s)
| | - C Michael DiPersio
- Department of Surgery, Albany Medical College, Albany, New York, United States
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, United States
| |
Collapse
|
5
|
Mishra J, Chakraborty S, Niharika, Roy A, Manna S, Baral T, Nandi P, Patra SK. Mechanotransduction and epigenetic modulations of chromatin: Role of mechanical signals in gene regulation. J Cell Biochem 2024; 125:e30531. [PMID: 38345428 DOI: 10.1002/jcb.30531] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 01/08/2024] [Accepted: 01/26/2024] [Indexed: 03/12/2024]
Abstract
Mechanical forces may be generated within a cell due to tissue stiffness, cytoskeletal reorganization, and the changes (even subtle) in the cell's physical surroundings. These changes of forces impose a mechanical tension within the intracellular protein network (both cytosolic and nuclear). Mechanical tension could be released by a series of protein-protein interactions often facilitated by membrane lipids, lectins and sugar molecules and thus generate a type of signal to drive cellular processes, including cell differentiation, polarity, growth, adhesion, movement, and survival. Recent experimental data have accentuated the molecular mechanism of this mechanical signal transduction pathway, dubbed mechanotransduction. Mechanosensitive proteins in the cell's plasma membrane discern the physical forces and channel the information to the cell interior. Cells respond to the message by altering their cytoskeletal arrangement and directly transmitting the signal to the nucleus through the connection of the cytoskeleton and nucleoskeleton before the information despatched to the nucleus by biochemical signaling pathways. Nuclear transmission of the force leads to the activation of chromatin modifiers and modulation of the epigenetic landscape, inducing chromatin reorganization and gene expression regulation; by the time chemical messengers (transcription factors) arrive into the nucleus. While significant research has been done on the role of mechanotransduction in tumor development and cancer progression/metastasis, the mechanistic basis of force-activated carcinogenesis is still enigmatic. Here, in this review, we have discussed the various cues and molecular connections to better comprehend the cellular mechanotransduction pathway, and we also explored the detailed role of some of the multiple players (proteins and macromolecular complexes) involved in mechanotransduction. Thus, we have described an avenue: how mechanical stress directs the epigenetic modifiers to modulate the epigenome of the cells and how aberrant stress leads to the cancer phenotype.
Collapse
Affiliation(s)
- Jagdish Mishra
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Subhajit Chakraborty
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Niharika
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Ankan Roy
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Soumen Manna
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Tirthankar Baral
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Piyasa Nandi
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Samir K Patra
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| |
Collapse
|
6
|
Devarajan R, Izzi V, Peltoketo H, Rask G, Kauppila S, Väisänen MR, Ruotsalainen H, Martínez-Nieto G, Karppinen SM, Väisänen T, Kaur I, Koivunen J, Sasaki T, Winqvist R, Manninen A, Wärnberg F, Sund M, Pihlajaniemi T, Heljasvaara R. Targeting collagen XVIII improves the efficiency of ErbB inhibitors in breast cancer models. J Clin Invest 2023; 133:e159181. [PMID: 37498672 DOI: 10.1172/jci159181] [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: 03/31/2022] [Accepted: 07/20/2023] [Indexed: 07/29/2023] Open
Abstract
The tumor extracellular matrix (ECM) critically regulates cancer progression and treatment response. Expression of the basement membrane component collagen XVIII (ColXVIII) is induced in solid tumors, but its involvement in tumorigenesis has remained elusive. We show here that ColXVIII was markedly upregulated in human breast cancer (BC) and was closely associated with a poor prognosis in high-grade BCs. We discovered a role for ColXVIII as a modulator of epidermal growth factor receptor tyrosine kinase (ErbB) signaling and show that it forms a complex with ErbB1 and -2 (also known as EGFR and human epidermal growth factor receptor 2 [HER2]) and α6-integrin to promote cancer cell proliferation in a pathway involving its N-terminal portion and the MAPK/ERK1/2 and PI3K/AKT cascades. Studies using Col18a1 mouse models crossed with the mouse mammary tumor virus-polyoma virus middle T antigen (MMTV-PyMT) mammary carcinogenesis model showed that ColXVIII promoted BC growth and metastasis in a tumor cell-autonomous manner. Moreover, the number of mammary cancer stem cells was significantly reduced in the MMTV-PyMT and human cell models upon ColXVIII inhibition. Finally, ablation of ColXVIII substantially improved the efficacy of ErbB-targeting therapies in both preclinical models. In summary, ColXVIII was found to sustain the stemness properties of BC cells and tumor progression and metastasis through ErbB signaling, suggesting that targeting ColXVIII in the tumor milieu may have important therapeutic potential.
Collapse
Affiliation(s)
- Raman Devarajan
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine
- Laboratory of Cancer Genetics and Tumor Biology, Cancer and Translational Medicine Research Unit
- Biocenter Oulu, and
| | - Valerio Izzi
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine
- Research Unit of Biomedicine, University of Oulu, Oulu, Finland
- Finnish Cancer Research Institute, Helsinki, Finland
| | - Hellevi Peltoketo
- Laboratory of Cancer Genetics and Tumor Biology, Cancer and Translational Medicine Research Unit
- Biocenter Oulu, and
| | - Gunilla Rask
- Department of Medical Biosciences/Pathology, Umeå University, Umeå, Sweden
| | - Saila Kauppila
- Department of Pathology, Oulu University Hospital and University of Oulu, Oulu, Finland
- Northern Finland Laboratory Centre, NordLab, Oulu, Finland
| | | | - Heli Ruotsalainen
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine
| | | | - Sanna-Maria Karppinen
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine
| | - Timo Väisänen
- Department of Pathology, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Inderjeet Kaur
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine
| | - Jussi Koivunen
- Department of Medical Oncology and Radiotherapy and Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Takako Sasaki
- Department of Pharmacology, Faculty of Medicine, Oita University, Oita, Japan
| | - Robert Winqvist
- Laboratory of Cancer Genetics and Tumor Biology, Cancer and Translational Medicine Research Unit
- Biocenter Oulu, and
- Northern Finland Laboratory Centre, NordLab, Oulu, Finland
| | - Aki Manninen
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Fredrik Wärnberg
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Malin Sund
- Department of Surgery and Perioperative Sciences/Surgery, Umeå University, Umeå, Sweden
- Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Taina Pihlajaniemi
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine
| | - Ritva Heljasvaara
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine
| |
Collapse
|
7
|
Tvaroška I, Kozmon S, Kóňa J. Molecular Modeling Insights into the Structure and Behavior of Integrins: A Review. Cells 2023; 12:cells12020324. [PMID: 36672259 PMCID: PMC9856412 DOI: 10.3390/cells12020324] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Integrins are heterodimeric glycoproteins crucial to the physiology and pathology of many biological functions. As adhesion molecules, they mediate immune cell trafficking, migration, and immunological synapse formation during inflammation and cancer. The recognition of the vital roles of integrins in various diseases revealed their therapeutic potential. Despite the great effort in the last thirty years, up to now, only seven integrin-based drugs have entered the market. Recent progress in deciphering integrin functions, signaling, and interactions with ligands, along with advancement in rational drug design strategies, provide an opportunity to exploit their therapeutic potential and discover novel agents. This review will discuss the molecular modeling methods used in determining integrins' dynamic properties and in providing information toward understanding their properties and function at the atomic level. Then, we will survey the relevant contributions and the current understanding of integrin structure, activation, the binding of essential ligands, and the role of molecular modeling methods in the rational design of antagonists. We will emphasize the role played by molecular modeling methods in progress in these areas and the designing of integrin antagonists.
Collapse
Affiliation(s)
- Igor Tvaroška
- Institute of Chemistry, Slovak Academy of Sciences, Dúbravska cesta 9, 845 38 Bratislava, Slovakia
- Correspondence:
| | - Stanislav Kozmon
- Institute of Chemistry, Slovak Academy of Sciences, Dúbravska cesta 9, 845 38 Bratislava, Slovakia
- Medical Vision o. z., Záhradnícka 4837/55, 821 08 Bratislava, Slovakia
| | - Juraj Kóňa
- Institute of Chemistry, Slovak Academy of Sciences, Dúbravska cesta 9, 845 38 Bratislava, Slovakia
- Medical Vision o. z., Záhradnícka 4837/55, 821 08 Bratislava, Slovakia
| |
Collapse
|
8
|
Chen JR, Zhao JT, Xie ZZ. Integrin-mediated cancer progression as a specific target in clinical therapy. Biomed Pharmacother 2022; 155:113745. [DOI: 10.1016/j.biopha.2022.113745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/17/2022] [Accepted: 09/21/2022] [Indexed: 11/15/2022] Open
|
9
|
Sui Y, Liu J, Zhang J, Zheng Z, Wang Z, Jia Z, Meng Z. Expression and Gene Regulation Network of Adenosine Receptor A2B in Lung Adenocarcinoma: A Potential Diagnostic and Prognostic Biomarker. Front Mol Biosci 2021; 8:663011. [PMID: 34350210 PMCID: PMC8326519 DOI: 10.3389/fmolb.2021.663011] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 06/17/2021] [Indexed: 12/25/2022] Open
Abstract
Adenosinereceptor A2B (ADORA2B) encodes a protein belonging to the G protein–coupled receptor superfamily. Abnormal expression of ADORA2B may play a pathophysiological role in some human cancers. We investigated whether ADORA2B is a potential diagnostic and prognostic biomarker for lung adenocarcinoma (LUAD). The expression, various mutations, copy number variations, mRNA expression levels, and related network signaling pathways of ADORA2B were analyzed using bioinformatics-related websites, including Oncomine, UALCAN, cBioPortal, GeneMANIA, LinkedOmics, KM Plotter, and TIMER. We found that ADORA2B was overexpressed and amplified in LUAD, and a high ADORA2B expression predicted a poor prognosis for LUAD patients. Pathway analyses of ADORA2B in LUAD revealed ADORA2B-correlated signaling pathways, and the expression level of ADORA2B was associated with immune cell infiltration. Furthermore, ADORA2B mRNA and protein levels were significantly higher in human LUAD cell lines (A549 cells and NCl-H1299 cells) than in normal human bronchial epithelial (HBE) cells, and the transcript levels of genes positively or negatively correlated with ADORA2B were consistent and statistically significant. siRNA transfection experiments and functional experiments further confirmed these results. In vitro results were also consistent with those of bioinformatics analysis. Our findings provide a foundation for studying the role of ADORA2B in tumorigenesis and support the development of new drug targets for LUAD.
Collapse
Affiliation(s)
- Yutong Sui
- Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Jiayin Liu
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jing Zhang
- Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Zena Zheng
- Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Ziwei Wang
- Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Zhenghu Jia
- The First Affiliated Hospital, Biomedical Translational Research Institute and Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China
| | - Ziyu Meng
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
10
|
Dzobo K. Integrins Within the Tumor Microenvironment: Biological Functions, Importance for Molecular Targeting, and Cancer Therapeutics Innovation. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2021; 25:417-430. [PMID: 34191612 DOI: 10.1089/omi.2021.0069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Many cellular functions important for solid tumor initiation and progression are mediated by members of the integrin family, a diverse family of cell attachment receptors. With recent studies emphasizing the role of the tumor microenvironment (TME) in tumor initiation and progression, it is not surprising that considerable attention is being paid to integrins. Several integrin antagonists are under clinical trials, with many demonstrating promising activity in patients with different cancers. A deeper knowledge of the functions of integrins within the TME is still required and might lead to better inhibitors being discovered. Integrin expression is commonly dysregulated in many tumors with integrins playing key roles in signaling as well as promotion of tumor cell invasion and migration. Integrins also play a major role in adhesion of circulating tumor cells to new sites and the resulting formation of secondary tumors. Furthermore, integrins have demonstrated the ability to promoting stem cell-like properties in tumor cells as well as drug resistance. Anti-integrin therapies rely heavily on the doses or concentrations used as these determine whether the drugs act as antagonists or as integrin agonists. This expert review offers the latest synthesis in terms of the current knowledge of integrins functions within the TME and as potential molecular targets for cancer therapeutics innovation.
Collapse
Affiliation(s)
- Kevin Dzobo
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa.,Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
11
|
Understanding the role of integrins in breast cancer invasion, metastasis, angiogenesis, and drug resistance. Oncogene 2021; 40:1043-1063. [PMID: 33420366 DOI: 10.1038/s41388-020-01588-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 11/11/2020] [Accepted: 11/26/2020] [Indexed: 12/13/2022]
Abstract
Integrins are cell adhesion receptors, which are typically transmembrane glycoproteins that connect to the extracellular matrix (ECM). The function of integrins regulated by biochemical events within the cells. Understanding the mechanisms of cell growth by integrins is important in elucidating their effects on tumor progression. One of the major events in integrin signaling is integrin binding to extracellular ligands. Another event is distant signaling that gathers chemical signals from outside of the cell and transmit the signals upon cell adhesion to the inside of the cell. In normal breast tissue, integrins function as checkpoints to monitor effects on cell proliferation, while in cancer tissue these functions altered. The combination of tumor microenvironment and its associated components determines the cell fate. Hypoxia can increase the expression of several integrins. The exosomal integrins promote the growth of metastatic cells. Expression of certain integrins is associated with increased metastasis and decreased prognosis in cancers. In addition, integrin-binding proteins promote invasion and metastasis in breast cancer. Targeting specific integrins and integrin-binding proteins may provide new therapeutic approaches for breast cancer therapies. This review will examine the current knowledge of integrins' role in breast cancer.
Collapse
|
12
|
Su CY, Li JQ, Zhang LL, Wang H, Wang FH, Tao YW, Wang YQ, Guo QR, Li JJ, Liu Y, Yan YY, Zhang JY. The Biological Functions and Clinical Applications of Integrins in Cancers. Front Pharmacol 2020; 11:579068. [PMID: 33041823 PMCID: PMC7522798 DOI: 10.3389/fphar.2020.579068] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/24/2020] [Indexed: 12/12/2022] Open
Abstract
Integrins are the adhesion molecules and receptors of extracellular matrix (ECM). They mediate the interactions between cells-cells and cells-ECM. The crosstalk between cancer cells and their microenvironment triggers a variety of critical signaling cues and promotes the malignant phenotype of cancer. As a type of transmembrane protein, integrin-mediated cell adhesion is essential in regulating various biological functions of cancer cells. Recent evidence has shown that integrins present on tumor cells or tumor-associated stromal cells are involved in ECM remodeling, and as mechanotransducers sensing changes in the biophysical properties of the ECM, which contribute to cancer metastasis, stemness and drug resistance. In this review, we outline the mechanism of integrin-mediated effects on biological changes of cancers and highlight the current status of clinical treatments by targeting integrins.
Collapse
Affiliation(s)
- Chao-yue Su
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jing-quan Li
- The First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Ling-ling Zhang
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Hui Wang
- Guangzhou Institute of Pediatrics/Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Feng-hua Wang
- Guangzhou Institute of Pediatrics/Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yi-wen Tao
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yu-qing Wang
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Qiao-ru Guo
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jia-jun Li
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yun Liu
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yan-yan Yan
- Institute of Immunology and School of Medicine, Shanxi Datong University, Datong, China
| | - Jian-ye Zhang
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
- The First Affiliated Hospital, Hainan Medical University, Haikou, China
| |
Collapse
|
13
|
Wenzel J, Rose K, Haghighi EB, Lamprecht C, Rauen G, Freihen V, Kesselring R, Boerries M, Hecht A. Loss of the nuclear Wnt pathway effector TCF7L2 promotes migration and invasion of human colorectal cancer cells. Oncogene 2020; 39:3893-3909. [PMID: 32203164 PMCID: PMC7203011 DOI: 10.1038/s41388-020-1259-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 12/25/2022]
Abstract
The transcription factor TCF7L2 is indispensable for intestinal tissue homeostasis where it transmits mitogenic Wnt/β-Catenin signals in stem and progenitor cells, from which intestinal tumors arise. Yet, TCF7L2 belongs to the most frequently mutated genes in colorectal cancer (CRC), and tumor-suppressive functions of TCF7L2 were proposed. This apparent paradox warrants to clarify the role of TCF7L2 in colorectal carcinogenesis. Here, we investigated TCF7L2 dependence/independence of CRC cells and the cellular and molecular consequences of TCF7L2 loss-of-function. By genome editing we achieved complete TCF7L2 inactivation in several CRC cell lines without loss of viability, showing that CRC cells have widely lost the strict requirement for TCF7L2. TCF7L2 deficiency impaired G1/S progression, reminiscent of the physiological role of TCF7L2. In addition, TCF7L2-negative cells exhibited morphological changes, enhanced migration, invasion, and collagen adhesion, albeit the severity of the phenotypic alterations manifested in a cell-line-specific fashion. To provide a molecular framework for the observed cellular changes, we performed global transcriptome profiling and identified gene-regulatory networks in which TCF7L2 positively regulates the proto-oncogene MYC, while repressing the cell cycle inhibitors CDKN2C/CDKN2D. Consistent with its function in curbing cell motility and invasion, TCF7L2 directly suppresses the pro-metastatic transcription factor RUNX2 and impinges on the expression of cell adhesion molecules. Altogether, we conclude that the proliferation-stimulating activity of TCF7L2 persists in CRC cells. In addition, TCF7L2 acts as invasion suppressor. Despite its negative impact on cell cycle progression, TCF7L2 loss-of-function may thereby increase malignancy, which could explain why TCF7L2 is mutated in a sizeable fraction of colorectal tumors.
Collapse
Affiliation(s)
- Janna Wenzel
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Stefan-Meier-Str. 17, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
| | - Katja Rose
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Stefan-Meier-Str. 17, 79104, Freiburg, Germany
| | - Elham Bavafaye Haghighi
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Breisacherstr. 153, 79104, Freiburg, Germany
| | - Constanze Lamprecht
- Institute of Physics, University of Freiburg, Hermann-Herder-Str. 3a, 79104, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Freiburg Center for Interactive Materials and Bioinspired Technology (FIT), University of Freiburg, Georges-Köhler-Allee 105, 79110, Freiburg, Germany
| | - Gilles Rauen
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
| | - Vivien Freihen
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Stefan-Meier-Str. 17, 79104, Freiburg, Germany
| | - Rebecca Kesselring
- Department of General and Visceral Surgery, Center for Surgery, Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Breisacherstr. 153, 79104, Freiburg, Germany
- German Cancer Consortium (DKTK), Hugstetter Straße 55, 79106, Freiburg, Germany
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Andreas Hecht
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Stefan-Meier-Str. 17, 79104, Freiburg, Germany.
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg, Germany.
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany.
| |
Collapse
|
14
|
Li Q, Peng K, Chen E, Jiang H, Wang Y, Yu S, Li W, Yu Y, Liu T. IntegrinB5 upregulated by HER2 in gastric cancer: a promising biomarker for liver metastasis. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:451. [PMID: 32395495 PMCID: PMC7210205 DOI: 10.21037/atm.2020.03.184] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 03/09/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Liver is the most frequent metastatic site of gastric cancer (GC), especially in patients with HER2 positive GC. Exosomal integrin αvβ5 has been shown to promote liver metastasis (LM), and the cross talk between integrins and HER2 during breast cancer metastasis has been reported. However, whether there's an association between HER2 and integrin αvβ5 (ITGAvB5), and whether their association has predictive value in GC liver metastasis (GCLM) remains unknown. METHODS The association between ITGβ5 and HER2 were accessed by RT-PCR, western blot and ELISA. We tested the function of ITGβ5 on HER2 positive GC cells using Transwell assays and scratch assays. Besides, we detect ITGβ5 expression in tumor tissue of GC patients and exosomes derived from advanced GC to analyze the association between HER2 and LM. RESULTS In our study, we found that ITGβ5, rather than ITGAV, was highly upregulated by HER2 through PI3K-AKT pathways in HER2 positive GC. Overexpression of ITGβ5 promoted the migration and invasion of HER2 positive GC cells in vitro. ITGβ5 was found to be an independent prognostic factor for GC. Besides, ITGβ5 level was only associated with LM. Detection of exosomal ITGβ5 and HER2 in the serum of GC patients revealed that exosomal ITGβ5 and HER2 levels are in accordance with that in tissue, and exosomal ITGβ5 level was higher in GCLM than other metastasis. CONCLUSIONS Our study demonstrated ITGβ5 is regulated and functions in accordance with HER2 in promoting GCLM. Exosomal ITGβ5 levels might be a potential liquid biopsy biomarker for GCLM.
Collapse
Affiliation(s)
- Qian Li
- Department of Medical Oncology, Zhongshan Hospital, and Center of Evidence-Based Medicine, Fudan University, Shanghai 200032, China
| | - Ke Peng
- Department of Medical Oncology, Zhongshan Hospital, and Center of Evidence-Based Medicine, Fudan University, Shanghai 200032, China
| | - Erbao Chen
- Department of Medical Oncology, Zhongshan Hospital, and Center of Evidence-Based Medicine, Fudan University, Shanghai 200032, China
| | - Huiqin Jiang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yan Wang
- Department of Medical Oncology, Zhongshan Hospital, and Center of Evidence-Based Medicine, Fudan University, Shanghai 200032, China
| | - Shan Yu
- Department of Medical Oncology, Zhongshan Hospital, and Center of Evidence-Based Medicine, Fudan University, Shanghai 200032, China
| | - Wei Li
- Department of Medical Oncology, Zhongshan Hospital, and Center of Evidence-Based Medicine, Fudan University, Shanghai 200032, China
| | - Yiyi Yu
- Department of Medical Oncology, Zhongshan Hospital, and Center of Evidence-Based Medicine, Fudan University, Shanghai 200032, China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, and Center of Evidence-Based Medicine, Fudan University, Shanghai 200032, China
| |
Collapse
|
15
|
The Extracellular Matrix: An Accomplice in Gastric Cancer Development and Progression. Cells 2020; 9:cells9020394. [PMID: 32046329 PMCID: PMC7072625 DOI: 10.3390/cells9020394] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/03/2020] [Accepted: 02/06/2020] [Indexed: 02/07/2023] Open
Abstract
The extracellular matrix (ECM) is a dynamic and highly organized tissue structure, providing support and maintaining normal epithelial architecture. In the last decade, increasing evidence has emerged demonstrating that alterations in ECM composition and assembly strongly affect cellular function and behavior. Even though the detailed mechanisms underlying cell-ECM crosstalk are yet to unravel, it is well established that ECM deregulation accompanies the development of many pathological conditions, such as gastric cancer. Notably, gastric cancer remains a worldwide concern, representing the third most frequent cause of cancer-associated deaths. Despite increased surveillance protocols, patients are usually diagnosed at advanced disease stages, urging the identification of novel diagnostic biomarkers and efficient therapeutic strategies. In this review, we provide a comprehensive overview regarding expression patterns of ECM components and cognate receptors described in normal gastric epithelium, pre-malignant lesions, and gastric carcinomas. Important insights are also discussed for the use of ECM-associated molecules as predictive biomarkers of the disease or as potential targets in gastric cancer.
Collapse
|
16
|
Extracellular matrix-cell interactions: Focus on therapeutic applications. Cell Signal 2019; 66:109487. [PMID: 31778739 DOI: 10.1016/j.cellsig.2019.109487] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/25/2019] [Accepted: 11/25/2019] [Indexed: 02/06/2023]
Abstract
Extracellular matrix (ECM) macromolecules together with a multitude of different molecules residing in the extracellular space play a vital role in the regulation of cellular phenotype and behavior. This is achieved via constant reciprocal interactions between the molecules of the ECM and the cells. The ECM-cell interactions are mediated via cell surface receptors either directly or indirectly with co-operative molecules. The ECM is also under perpetual remodeling process influencing cell-signaling pathways on its part. The fragmentation of ECM macromolecules provides even further complexity for the intricate environment of the cells. However, as long as the interactions between the ECM and the cells are in balance, the health of the body is retained. Alternatively, any dysregulation in these interactions can lead to pathological processes and finally to various diseases. Thus, therapeutic applications that are based on retaining normal ECM-cell interactions are highly rationale. Moreover, in the light of the current knowledge, also concurrent multi-targeting of the complex ECM-cell interactions is required for potent pharmacotherapies to be developed in the future.
Collapse
|