1
|
Huang Q, Wang J, Ning H, Liu W, Han X. Integrin β1 in breast cancer: mechanisms of progression and therapy. Breast Cancer 2024:10.1007/s12282-024-01635-w. [PMID: 39343856 DOI: 10.1007/s12282-024-01635-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024]
Abstract
The therapy for breast cancer (BC), to date, still needs improvement. Apart from traditional therapy methods, biological therapy being explored opens up a novel avenue for BC patients. Integrin β1 (ITGβ1), one of the largest subgroups in integrin family, is a key player in cancer evolution and therapy. Recent researches progress in the relationship of ITGβ1 level and BC, finding that ITGβ1 expression evidently concerns BC progression. In this chapter, we outline diverse ITGβ1-based mechanisms regarding to the promoted effect of ITGβ1 on BC cell structure rearrangement and malignant phenotype behaviors, the unfavorable patient prognosis conferred by ITGβ1, BC therapy tolerance induced by ITGβ1, and lastly novel inhibitors targeting ITGβ1 for BC therapy. As an effective biomarker, ITGβ1 undoubtedly emerges one of targeted-therapy opportunities of BC patients in future. It is a necessity focusing on scientific and large-scale clinical trials on the validation of targeted-ITGβ1 drugs for BC patients.
Collapse
Affiliation(s)
- Qionglian Huang
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jue Wang
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hanjuan Ning
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weiwei Liu
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xianghui Han
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
2
|
Malla R, Jyosthsna K, Rani G, Purnachandra Nagaraju G. CD44/PD-L1-mediated networks in drug resistance and immune evasion of breast cancer stem cells: Promising targets of natural compounds. Int Immunopharmacol 2024; 138:112613. [PMID: 38959542 DOI: 10.1016/j.intimp.2024.112613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 06/28/2024] [Accepted: 06/30/2024] [Indexed: 07/05/2024]
Abstract
Cancer stem cells (CSCs) significantly interfere with immunotherapy, leading to challenges such as low response rates and acquired resistance. PD-L1 expression is associated with the CSC population's overexpression of CD44. Mounting evidence suggests that the breast cancer stem cell (BCSC) marker CD44 and the immune checkpoint PD-L1 contribute to treatment failure through their networks. Natural compounds can overcome therapy resistance in breast cancer by targeting mechanisms underlying resistance in BCSCs. This review provides an updated insight into the CD44 and PD-L1 networks of BCSCs in mediating metastasis and immune evasion. The review critically examines existing literature, providing a comprehensive understanding of the topic and emphasizing the impact of natural flavones on the signaling pathways of BCSCs. Additionally, the review discusses the potential of natural compounds in targeting CD44 and PD-L1 in breast cancer (BC). Natural compounds consistently show potential in targeting regulatory mechanisms of BCSCs, inducing loss of stemness, and promoting differentiation. They offer a promising approach for developing alternative therapeutic strategies to manage breast cancer.
Collapse
Affiliation(s)
- RamaRao Malla
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India; Department of Biochemistry and Bioinformatics, School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India.
| | - Kattula Jyosthsna
- Department of Biotechnology, School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India
| | - G Rani
- Department of Biotechnology, School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35233, USA
| |
Collapse
|
3
|
Hu Q, Remsing Rix LL, Desai B, Miroshnychenko D, Li X, Welsh EA, Fang B, Wright GM, Chaudhary N, Kroeger JL, Doebele RC, Koomen JM, Haura EB, Marusyk A, Rix U. Cancer-associated fibroblasts confer ALK inhibitor resistance in EML4-ALK -driven lung cancer via concurrent integrin and MET signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.27.609975. [PMID: 39253447 PMCID: PMC11383036 DOI: 10.1101/2024.08.27.609975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Cancer-associated fibroblasts (CAFs) are associated with tumor progression and modulate drug sensitivity of cancer cells. However, the underlying mechanisms are often incompletely understood and crosstalk between tumor cells and CAFs involves soluble secreted as well as adhesion proteins. Interrogating a panel of non-small cell lung cancer (NSCLC) cell lines driven by EML4-ALK fusions, we observed substantial CAF-mediated drug resistance to clinical ALK tyrosine kinase inhibitors (TKIs). Array-based cytokine profiling of fibroblast-derived conditioned- media identified HGF-MET signaling as a major contributor to CAF-mediated paracrine resistance that can be overcome by MET TKIs. However, 'Cell Type specific labeling using Amino acid Precursors' (CTAP)-based expression and phosphoproteomics in direct coculture also highlighted a critical role for the fibronectin-integrin pathway. Flow cytometry analysis confirmed activation of integrin β1 (ITGB1) in lung cancer cells by CAF coculture. Treatment with pharmacological inhibitors, cancer cell-specific silencing or CRISPR-Cas9-mediated knockout of ITGB1 overcame adhesion protein-mediated resistance. Concurrent targeting of MET and integrin signaling effectively abrogated CAF-mediated resistance of EML4-ALK -driven NSCLC cells to ALK TKIs in vitro . Consistently, combination of the ALK TKI alectinib with the MET TKI capmatinib and/or the integrin inhibitor cilengitide was significantly more efficacious than single agent treatment in suppressing tumor growth using an in vivo EML4-ALK -dependent allograft mouse model of NSCLC. In summary, these findings emphasize the complexity of resistance-associated crosstalk between CAFs and cancer cells, which can involve multiple concurrent signaling pathways, and illustrate how comprehensive elucidation of paracrine and juxtacrine resistance mechanisms can inform on more effective therapeutic approaches.
Collapse
|
4
|
Cheung BCH, Chen X, Davis HJ, Nordmann CS, Toth J, Hodgson L, Segall JE, Shenoy VB, Wu M. Identification of CD44 as a key mediator of cell traction force generation in hyaluronic acid-rich extracellular matrices. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.24.563860. [PMID: 37961689 PMCID: PMC10634813 DOI: 10.1101/2023.10.24.563860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Mechanical properties of the extracellular matrix (ECM) critically regulate a number of important cell functions including growth, differentiation and migration. Type I collagen and glycosaminoglycans (GAGs) are two primary components of ECMs that contribute to mammalian tissue mechanics, with the collagen fiber network sustaining tension, and GAGs withstanding compression. The architecture and stiffness of the collagen network are known to be important for cell-ECM mechanical interactions via integrin cell surface adhesion receptors. In contrast, studies of GAGs in modulating cell-ECM interactions are limited. Here, we present experimental studies on the roles of hyaluronic acid (HA, an unsulfated GAG) in single tumor cell traction force generation using a recently developed 3D cell traction force microscopy method. Our work reveals that CD44, a cell surface adhesion receptor to HA, is engaged in cell traction force generation in conjunction with β1-integrin. We find that HA significantly modifies the architecture and mechanics of the collagen fiber network, decreasing tumor cells' propensity to remodel the collagen network, attenuating traction force generation, transmission distance, and tumor invasion. Our findings point to a novel role for CD44 in traction force generation, which can be a potential therapeutic target for diseases involving HA rich ECMs such as breast cancer and glioblastoma.
Collapse
|
5
|
Behl T, Kumar A, Vishakha, Sehgal A, Singh S, Sharma N, Yadav S, Rashid S, Ali N, Ahmed AS, Vargas-De-La-Cruz C, Bungau SG, Khan H. Understanding the mechanistic pathways and clinical aspects associated with protein and gene based biomarkers in breast cancer. Int J Biol Macromol 2023; 253:126595. [PMID: 37648139 DOI: 10.1016/j.ijbiomac.2023.126595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/22/2023] [Accepted: 08/27/2023] [Indexed: 09/01/2023]
Abstract
Cancer is one of the most widespread and severe diseases with a huge mortality rate. In recent years, the second-leading mortality rate of any cancer globally has been breast cancer, which is one of the most common and deadly cancers found in women. Detecting breast cancer in its initial stages simplifies treatment, decreases death risk, and recovers survival rates for patients. The death rate for breast cancer has risen to 0.024 % in some regions. Sensitive and accurate technologies are required for the preclinical detection of BC at an initial stage. Biomarkers play a very crucial role in the early identification as well as diagnosis of women with breast cancer. Currently, a wide variety of cancer biomarkers have been discovered for the diagnosis of cancer. For the identification of these biomarkers from serum or other body fluids at physiological amounts, many detection methods have been developed. In the case of breast cancer, biomarkers are especially helpful in discovering those who are more likely to develop the disease, determining prognosis at the time of initial diagnosis and choosing the best systemic therapy. In this study we have compiled various clinical aspects and signaling pathways associated with protein-based biomarkers and gene-based biomarkers.
Collapse
Affiliation(s)
- Tapan Behl
- School of Health Sciences and Technology, University of Petroleum and Energy Studies, Dehradun 248007, Uttarakhand, India
| | - Ankush Kumar
- Institute of Pharmaceutical Sciences, IET Bhaddal Technical Campus, Ropar 140108, Punjab, India
| | - Vishakha
- Institute of Pharmaceutical Sciences, IET Bhaddal Technical Campus, Ropar 140108, Punjab, India
| | - Aayush Sehgal
- GHG Khalsa College of Pharmacy, Gurusar Sadhar, 141104 Ludhiana, Punjab, India
| | - Sukhbir Singh
- Department of Pharmaceutics, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana Ambala 133203, Haryana, India
| | - Neelam Sharma
- Department of Pharmaceutics, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana Ambala 133203, Haryana, India
| | - Shivam Yadav
- School of Pharmacy, Babu Banarasi Das University, Lucknow 226028, Uttar Pradesh, India
| | - Summya Rashid
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia.
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadah 11451, Saudi Arabia
| | - Amira Saber Ahmed
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Giza 12622, Egypt
| | - Celia Vargas-De-La-Cruz
- Department of Pharmacology, Bromatology and Toxicology, Faculty of Pharmacy and Biochemistry, Universidad Nacional Mayor de San Marcos, Lima 150001, Peru; E-Health Research Center, Universidad de Ciencias y Humanidades, Lima 15001, Peru
| | - Simona Gabriela Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea 410087, Romania; Doctoral School of Biomedical Sciences, University of Oradea, Oradea 410087, Romania
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan.
| |
Collapse
|
6
|
Rist D, DePalma T, Stagner E, Tallman MM, Venere M, Skardal A, Schultz ZD. Cancer Cell Targeting, Magnetic Sorting, and SERS Detection through Cell Surface Receptors. ACS Sens 2023; 8:4636-4645. [PMID: 37988612 PMCID: PMC10921760 DOI: 10.1021/acssensors.3c01625] [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] [Indexed: 11/23/2023]
Abstract
Integrins are cellular surface receptors responsible for the activation of many cellular pathways in cancer. These integrin proteins can be specifically targeted by small peptide sequences that offer the potential for the differentiation of cellular subpopulations by using magnetically assisted cellular sorting techniques. By adding a gold shell to the magnetic nanoparticles, these integrin-peptide interactions can be differentiated by surface-enhanced Raman spectroscopy (SERS), providing a quick and reliable method for on-target binding. In this paper, we demonstrate the ability to differentiate the peptide-protein interactions of the small peptides CDPGYIGSR and cyclic RGDfC functionalized on gold-coated magnetic nanoparticles with the integrins they are known to bind to using their SERS signal. SW480 and SW620 colorectal cancer cells known to have the integrins of interest were then magnetically sorted using these functionalized nanoparticles, suggesting differentiation between the sorted populations and integrin populations among the two cell lines.
Collapse
Affiliation(s)
- David Rist
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, United States
| | - Tom DePalma
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Emerie Stagner
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Miranda M Tallman
- Department of Radiation Oncology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Monica Venere
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
- Department of Radiation Oncology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Aleksander Skardal
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Zachary D. Schultz
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, United States
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
7
|
Conway JRW, Isomursu A, Follain G, Härmä V, Jou-Ollé E, Pasquier N, Välimäki EPO, Rantala JK, Ivaska J. Defined extracellular matrix compositions support stiffness-insensitive cell spreading and adhesion signaling. Proc Natl Acad Sci U S A 2023; 120:e2304288120. [PMID: 37844244 PMCID: PMC10614832 DOI: 10.1073/pnas.2304288120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 09/15/2023] [Indexed: 10/18/2023] Open
Abstract
Integrin-dependent adhesion to the extracellular matrix (ECM) mediates mechanosensing and signaling in response to altered microenvironmental conditions. In order to provide tissue- and organ-specific cues, the ECM is composed of many different proteins that temper the mechanical properties and provide the necessary structural diversity. Despite most human tissues being soft, the prevailing view from predominantly in vitro studies is that increased stiffness triggers effective cell spreading and activation of mechanosensitive signaling pathways. To address the functional coupling of ECM composition and matrix rigidity on compliant substrates, we developed a matrix spot array system to screen cell phenotypes against different ECM mixtures on defined substrate stiffnesses at high resolution. We applied this system to both cancer and normal cells and surprisingly identified ECM mixtures that support stiffness-insensitive cell spreading on soft substrates. Employing the motor-clutch model to simulate cell adhesion on biochemically distinct soft substrates, with varying numbers of available ECM-integrin-cytoskeleton (clutch) connections, we identified conditions in which spreading would be supported on soft matrices. Combining simulations and experiments, we show that cell spreading on soft is supported by increased clutch engagement on specific ECM mixtures and even augmented by the partial inhibition of actomyosin contractility. Thus, "stiff-like" spreading on soft is determined by a balance of a cell's contractile and adhesive machinery. This provides a fundamental perspective for in vitro mechanobiology studies, identifying a mechanism through which cells spread, function, and signal effectively on soft substrates.
Collapse
Affiliation(s)
- James R. W. Conway
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, TurkuFI-20520, Finland
| | - Aleksi Isomursu
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, TurkuFI-20520, Finland
| | - Gautier Follain
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, TurkuFI-20520, Finland
| | - Ville Härmä
- Misvik Biology Oy, TurkuFI-20520, Finland
- Department of Oncology and Metabolism, University of Sheffield, SheffieldS10 2TN, United Kingdom
| | - Eva Jou-Ollé
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, TurkuFI-20520, Finland
| | - Nicolas Pasquier
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, TurkuFI-20520, Finland
| | | | - Juha K. Rantala
- Misvik Biology Oy, TurkuFI-20520, Finland
- Department of Oncology and Metabolism, University of Sheffield, SheffieldS10 2TN, United Kingdom
| | - Johanna Ivaska
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, TurkuFI-20520, Finland
- Department of Life Technologies, University of Turku, TurkuFI-20520, Finland
- InFLAMES Research Flagship, University of Turku, TurkuFI-20520, Finland
- Western Finnish Cancer Center, University of Turku, TurkuFI-20520, Finland
- Foundation for the Finnish Cancer Institute, HelsinkiFI-00014, Finland
| |
Collapse
|
8
|
Aguilar-Chaparro MA, Rivera-Pineda SA, Hernández-Galdámez HV, Piña-Vázquez C, Villa-Treviño S. The CD44std and CD44v9 subpopulations in non-tumorigenic invasive SNU-423 cells present different features of cancer stem cells. Stem Cell Res 2023; 72:103222. [PMID: 37844417 DOI: 10.1016/j.scr.2023.103222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/11/2023] [Accepted: 10/10/2023] [Indexed: 10/18/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a type of liver cancer, in which CD44 isoforms have been proposed as markers to identify cancer stem cells (CSCs). However, it is unclear what characteristics are associated with CSCs that exclusively express CD44 isoforms. The objective of the present study was to determine the expression of CD44 isoforms and their properties in CSCs. Analysis of transcriptomic data from HCC patient samples identified CD44v8-10 as a potential marker in HCC. In SNU-423 cells, CD44 expression was detected in over 99% of cells, and two CD44 isoforms, namely, CD44std and CD44v9, were identified in this cell line. CD44 subpopulations, including both CD44v9+ (CD44v9) and CD44v9- (CD44std) cells, were obtained by purification using a magnetic cell separation kit for human CD44v9+ cancer stem cells. CD44v9 cells showed greater potential for colony and spheroid formation, whereas CD44std cells demonstrated significant migration and invasion capabilities. These findings suggested that CD44std and CD44v9 may be used to identify features in CSC populations and provide insights into their roles in HCC.
Collapse
Affiliation(s)
- Mario Alejandro Aguilar-Chaparro
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV), Av. IPN No. 2508 Col. San Pedro Zacatenco, México City CP 07360, Mexico
| | - Sonia Andrea Rivera-Pineda
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV), Av. IPN No. 2508 Col. San Pedro Zacatenco, México City CP 07360, Mexico
| | - Hury Viridiana Hernández-Galdámez
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV), Av. IPN No. 2508 Col. San Pedro Zacatenco, México City CP 07360, Mexico
| | - Carolina Piña-Vázquez
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV), Av. IPN No. 2508 Col. San Pedro Zacatenco, México City CP 07360, Mexico
| | - Saúl Villa-Treviño
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV), Av. IPN No. 2508 Col. San Pedro Zacatenco, México City CP 07360, Mexico.
| |
Collapse
|
9
|
Savli TB, Pasaoglu HE, Savli TC, Muhammedoglu A, Tokocin M, Öztürk Ç. Expression of cytotoxic T lymphocyte-associated antigen 4, CD44, and E-cadherin in the microenvironment of breast carcinomas. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2023; 69:e20230371. [PMID: 37466609 PMCID: PMC10351999 DOI: 10.1590/1806-9282.20230371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 04/28/2023] [Indexed: 07/20/2023]
Abstract
OBJECTIVE The expression of cytotoxic T lymphocyte-associated antigen 4, E-cadherin, and CD44 in the area of tumor budding was investigated in breast carcinomas in our study. METHODS Tumor budding was counted at the invasive margins in 179 breast carcinomas. To understand the microenvironment of tumor budding, we examined the expression status of the immune checkpoint molecules such as cytotoxic T lymphocyte-associated antigen 4, E-cadherin, and CD44. RESULTS Tumors were separated into low (≤5) and high tumor budding groups (>5) based on the median budding number. Lymphovascular, perineural invasion, and the number of metastatic lymph nodes were significantly higher in high-grade budding tumors (p=0.001, p<0.001, and p=0.019, respectively). Tumor-infiltrating lymphocytes were significantly higher in tumors without tumor buddings (p<0.001). When the number of budding increases by one unit, overall survival decreases by 1.07 times (p=0.013). Also, it increases the risk of progression by 1.06 times (p=0.048). In high tumor budding groups, the cytotoxic T lymphocyte-associated antigen 4 staining percentage of lymphocytes was significantly higher (p=0.026). With each increase in the number of buds, an increase in the percentage of cytotoxic T lymphocyte-associated antigen 4 staining was seen in lymphocytes in the microenvironment of TB (p=0.034). CONCLUSION Tumor budding could predict poor prognosis in breast carcinomas, and anti-cytotoxic T lymphocyte-associated antigen 4 immunotherapies may be beneficial in patients with high tumor budding tumors.
Collapse
Affiliation(s)
- Tugce Bolme Savli
- Gaziantep Cengiz Gokcek Maternity and Child Health Hospital, Department of Pathology - Gaziantep, Turkey
| | - Husniye Esra Pasaoglu
- Istanbul Bagcilar Training and Research Hospital, Department of Pathology - Istanbul, Turkey
| | - Taha Cumhan Savli
- Medipol Mega University Hospital, Department of Pathology - Istanbul, Turkey
| | - Ali Muhammedoglu
- Istanbul Bagcilar Training and Research Hospital, Department of Pathology - Istanbul, Turkey
| | - Merve Tokocin
- Istanbul Bagcilar Training and Research Hospital, Department of General Surgery - Istanbul, Turkey
| | - Çiğdem Öztürk
- Recep Tayyip Erdoğan University Training and Research Hospital, Pathology Department - Rize, Turkey
| |
Collapse
|
10
|
Pawlak MR, Smiley AT, Ramirez MP, Kelly MD, Shamsan GA, Anderson SM, Smeester BA, Largaespada DA, Odde DJ, Gordon WR. RAD-TGTs: high-throughput measurement of cellular mechanotype via rupture and delivery of DNA tension probes. Nat Commun 2023; 14:2468. [PMID: 37117218 PMCID: PMC10147940 DOI: 10.1038/s41467-023-38157-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 04/19/2023] [Indexed: 04/30/2023] Open
Abstract
Mechanical forces drive critical cellular processes that are reflected in mechanical phenotypes, or mechanotypes, of cells and their microenvironment. We present here "Rupture And Deliver" Tension Gauge Tethers (RAD-TGTs) in which flow cytometry is used to record the mechanical history of thousands of cells exerting forces on their surroundings via their propensity to rupture immobilized DNA duplex tension probes. We demonstrate that RAD-TGTs recapitulate prior DNA tension probe studies while also yielding a gain of fluorescence in the force-generating cell that is detectable by flow cytometry. Furthermore, the rupture propensity is altered following disruption of the cytoskeleton using drugs or CRISPR-knockout of mechanosensing proteins. Importantly, RAD-TGTs can differentiate distinct mechanotypes among mixed populations of cells. We also establish oligo rupture and delivery can be measured via DNA sequencing. RAD-TGTs provide a facile and powerful assay to enable high-throughput mechanotype profiling, which could find various applications, for example, in combination with CRISPR screens and -omics analysis.
Collapse
Affiliation(s)
- Matthew R Pawlak
- Departments of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Adam T Smiley
- Departments of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Maria Paz Ramirez
- Departments of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Marcus D Kelly
- Departments of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Ghaidan A Shamsan
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Sarah M Anderson
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | | | | | - David J Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Wendy R Gordon
- Departments of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
11
|
Carvalho AM, Reis RL, Pashkuleva I. Hyaluronan Receptors as Mediators and Modulators of the Tumor Microenvironment. Adv Healthc Mater 2023; 12:e2202118. [PMID: 36373221 PMCID: PMC11469756 DOI: 10.1002/adhm.202202118] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/28/2022] [Indexed: 11/16/2022]
Abstract
The tumor microenvironment (TME) is a dynamic and complex matter shaped by heterogenous cancer and cancer-associated cells present at the tumor site. Hyaluronan (HA) is a major TME component that plays pro-tumorigenic and carcinogenic functions. These functions are mediated by different hyaladherins expressed by cancer and tumor-associated cells triggering downstream signaling pathways that determine cell fate and contribute to TME progression toward a carcinogenic state. Here, the interaction of HA is reviewed with several cell-surface hyaladherins-CD44, RHAMM, TLR2 and 4, LYVE-1, HARE, and layilin. The signaling pathways activated by these interactions and the respective response of different cell populations within the TME, and the modulation of the TME, are discussed. Potential cancer therapies via targeting these interactions are also briefly discussed.
Collapse
Affiliation(s)
- Ana M. Carvalho
- 3Bs Research Group, I3Bs ‐ Research Institute on Biomaterials Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineBarco4805‐017Portugal
- ICVS/3B's – PT Government Associate LaboratoryUniversity of MinhoBraga4710‐057Portugal
| | - Rui L. Reis
- 3Bs Research Group, I3Bs ‐ Research Institute on Biomaterials Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineBarco4805‐017Portugal
- ICVS/3B's – PT Government Associate LaboratoryUniversity of MinhoBraga4710‐057Portugal
| | - Iva Pashkuleva
- 3Bs Research Group, I3Bs ‐ Research Institute on Biomaterials Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineBarco4805‐017Portugal
- ICVS/3B's – PT Government Associate LaboratoryUniversity of MinhoBraga4710‐057Portugal
| |
Collapse
|
12
|
Moderate Constraint Facilitates Association and Force-Dependent Dissociation of HA-CD44 Complex. Int J Mol Sci 2023; 24:ijms24032243. [PMID: 36768572 PMCID: PMC9917194 DOI: 10.3390/ijms24032243] [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: 12/14/2022] [Revised: 01/10/2023] [Accepted: 01/21/2023] [Indexed: 01/25/2023] Open
Abstract
Binding of cell surface glycoprotein CD44 to hyaluronic acid (HA) is a key event for mediating cell adhesion, motility, metastasis, inflammatory responses and tumor development, but the regulation mechanism and its molecular basis under diverse mechanical constraints remain unclear. We herein investigated interaction of CD44 HABD (HA binding site domain) to HA through free and steered molecular dynamics (MD) simulations as well as atomic force microscope (AFM) measurement using different constraints on HA. The middle, two ends or both of the constrained HA chains were fixed for MD simulations, while one and two biotin-avidin linkage or physical absorption were used to immobilize HA on substrates for AFM experiments, to model HA chains with low, moderate and high HA flexibilities, respectively. We found that binding of CD44 to moderate fixed HA was possessed of a better thermo-stability, a lower mechanical strength and a higher dissociation probability, while higher adhesive frequency, smaller rupture force and shorter lifetime were assigned to CD44 on the two biotin-immobilized HA rather than one biotin-immobilized or physically absorbed HA on substrates, suggesting a moderate HA flexibility requirement in favor of association and force-induced dissociation of CD44-HA complex. Tensile-induced convex conformation of HA chain was responsible for reduction of complex mechano-stability and did inversely a shrunken CD44 HABD under stretching; transition from catch bond to slip bond governed CD44-HA interaction. This study uncovered the regulation mechanism and its molecular basis for CD44-HA affinity under diverse mechano-microenvironments and provided a new insight into CD44-HA interaction-mediated cell inflammatory responses and tumor development.
Collapse
|
13
|
Karagiorgou Z, Fountas PN, Manou D, Knutsen E, Theocharis AD. Proteoglycans Determine the Dynamic Landscape of EMT and Cancer Cell Stemness. Cancers (Basel) 2022; 14:5328. [PMID: 36358747 PMCID: PMC9653992 DOI: 10.3390/cancers14215328] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/23/2022] [Accepted: 10/25/2022] [Indexed: 03/15/2024] Open
Abstract
Proteoglycans (PGs) are pivotal components of extracellular matrices, involved in a variety of processes such as migration, invasion, morphogenesis, differentiation, drug resistance, and epithelial-to-mesenchymal transition (EMT). Cellular plasticity is a crucial intermediate phenotypic state acquired by cancer cells, which can modulate EMT and the generation of cancer stem cells (CSCs). PGs affect cell plasticity, stemness, and EMT, altering the cellular shape and functions. PGs control these functions, either by direct activation of signaling cascades, acting as co-receptors, or through regulation of the availability of biological compounds such as growth factors and cytokines. Differential expression of microRNAs is also associated with the expression of PGs and their interplay is implicated in the fine tuning of cancer cell phenotype and potential. This review summarizes the involvement of PGs in the regulation of EMT and stemness of cancer cells and highlights the molecular mechanisms.
Collapse
Affiliation(s)
- Zoi Karagiorgou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| | - Panagiotis N. Fountas
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| | - Dimitra Manou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| | - Erik Knutsen
- Department of Medical Biology, Faculty of Health Sciences, UiT the Arctic University of Norway, 9010 Tromsø, Norway
- Centre for Clinical Research and Education, University Hospital of North Norway, 9038 Tromsø, Norway
| | - Achilleas D. Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| |
Collapse
|
14
|
Carvalho AM, Soares da Costa D, Reis RL, Pashkuleva I. RHAMM expression tunes the response of breast cancer cell lines to hyaluronan. Acta Biomater 2022; 146:187-196. [PMID: 35577044 DOI: 10.1016/j.actbio.2022.05.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/12/2022] [Accepted: 05/08/2022] [Indexed: 02/06/2023]
Abstract
Hyaluronan (HA) synthesis and degradation are altered during carcinogenesis leading to an increased HA content in the tumor microenvironment, which correlates with poor prognosis and treatment outcomes. The main HA receptors, CD44 and RHAMM, are also overexpressed in tumors where they activate anti-apoptotic, proliferative, invasive, and migration signaling pathways. Herein, we used a unidirectional HA gradient to investigate in a high-throughput fashion the bi-directional communication between HA and breast cancer cell lines with different surface expression of CD44 and RHAMM. We found that the expression of CD44 and RHAMM depends on the HA density: the expression of these receptors is promoted at higher HA density and RHAMM is more sensitive to these changes when compared to CD44. Blocking either CD44 or RHAMM revealed different functions on binding and recognizing HA and a compensatory expression between these two receptors that maintains protumorigenic effectors such as cortactin. STATEMENT OF SIGNIFICANCE: We show that the expression of main hyaluronan (HA) receptors CD44 and RHAMM is enhanced in a HA concentration-dependent manner. Blocking activity experiments with either RHAMM or CD44 reveal the redundancy of these two receptors towards HA recognition and activation/recruitment of protumorigenic molecular effector, cortactin. These experiments also demonstrate that cells with overexpressed RHAMM are more sensitive to HA density than CD44 positive cells. The reported results are important for the development of therapies that target the hyaluronan signaling in the tumor microenvironment.
Collapse
Affiliation(s)
- Ana M Carvalho
- 3B's Research Group, I3Bs Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Barco, Portugal; ICVS/3B's - PT Government Associate Laboratory, University of Minho, Portugal
| | - Diana Soares da Costa
- 3B's Research Group, I3Bs Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Barco, Portugal; ICVS/3B's - PT Government Associate Laboratory, University of Minho, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Barco, Portugal; ICVS/3B's - PT Government Associate Laboratory, University of Minho, Portugal
| | - Iva Pashkuleva
- 3B's Research Group, I3Bs Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Barco, Portugal; ICVS/3B's - PT Government Associate Laboratory, University of Minho, Portugal.
| |
Collapse
|
15
|
Fernández-Tabanera E, Melero-Fernández de Mera RM, Alonso J. CD44 In Sarcomas: A Comprehensive Review and Future Perspectives. Front Oncol 2022; 12:909450. [PMID: 35785191 PMCID: PMC9247467 DOI: 10.3389/fonc.2022.909450] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/02/2022] [Indexed: 12/16/2022] Open
Abstract
It is widely accepted that the tumor microenvironment, particularly the extracellular matrix, plays an essential role in the development of tumors through the interaction with specific protein-membrane receptors. One of the most relevant proteins in this context is the transmembrane protein CD44. The role of CD44 in tumor progression, invasion, and metastasis has been well established in many cancers, although a comprehensive review concerning its role in sarcomas has not been published. CD44 is overexpressed in most sarcomas and several in vitro and in vivo experiments have shown a direct effect on tumor progression, dissemination, and drug resistance. Moreover, CD44 has been revealed as a useful marker for prognostic and diagnostic (CD44v6 isoform) in osteosarcoma. Besides, some innovative treatments such as HA-functionalized liposomes therapy have become an excellent CD44-mediated intracellular delivery system for osteosarcoma. Unfortunately, the reduced number of studies deciphering the prognostic/diagnostic value of CD44 in other sarcoma subgroups, neither than osteosarcoma, in addition to the low number of patients involved in those studies, have produced inconclusive results. In this review, we have gone through the information available on the role of CD44 in the development, maintenance, and progression of sarcomas, analyzing their implications at the prognostic, therapeutic, and mechanistic levels. Moreover, we illustrate how research involving the specific role of CD44 in the different sarcoma subgroups could suppose a chance to advance towards a more innovative perspective for novel therapies and future clinical trials.
Collapse
Affiliation(s)
- Enrique Fernández-Tabanera
- Unidad de Tumores Sólidos Infantiles, Instituto de Investigación de Enfermedades Raras (IIER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III (U758; CB06/07/1009; CIBERER-ISCIII), Madrid, Spain
- Universidad Nacional de Educación a Distancia (UNED), Madrid, Spain
| | - Raquel M. Melero-Fernández de Mera
- Unidad de Tumores Sólidos Infantiles, Instituto de Investigación de Enfermedades Raras (IIER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III (U758; CB06/07/1009; CIBERER-ISCIII), Madrid, Spain
| | - Javier Alonso
- Unidad de Tumores Sólidos Infantiles, Instituto de Investigación de Enfermedades Raras (IIER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III (U758; CB06/07/1009; CIBERER-ISCIII), Madrid, Spain
- *Correspondence: Javier Alonso,
| |
Collapse
|
16
|
DeCastro AJL, Pranda MA, Gray KM, Merlo-Coyne J, Girma N, Hurwitz M, Zhang Y, Stroka KM. Morphological Phenotyping of Organotropic Brain- and Bone-Seeking Triple Negative Metastatic Breast Tumor Cells. Front Cell Dev Biol 2022; 10:790410. [PMID: 35252171 PMCID: PMC8891987 DOI: 10.3389/fcell.2022.790410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/31/2022] [Indexed: 11/22/2022] Open
Abstract
Triple negative breast cancer (TNBC) follows a non-random pattern of metastasis to the bone and brain tissue. Prior work has found that brain-seeking breast tumor cells display altered proteomic profiles, leading to alterations in pathways related to cell signaling, cell cycle, metabolism, and extracellular matrix remodeling. Given the unique microenvironmental characteristics of brain and bone tissue, we hypothesized that brain- or bone-seeking TNBC cells may have altered morphologic or migratory phenotypes from each other, or from the parental TNBC cells, as a function of the biochemical or mechanical microenvironment. In this study, we utilized TNBC cells (MDA-MB-231) that were conditioned to metastasize solely to brain (MDA-BR) or bone (MDA-BO) tissue. We quantified characteristics such as cell morphology, migration, and stiffness in response to cues that partially mimic their final metastatic niche. We have shown that MDA-BO cells have a distinct protrusive morphology not found in MDA-P or MDA-BR. Further, MDA-BO cells migrate over a larger area when on a collagen I (abundant in bone tissue) substrate when compared to fibronectin (abundant in brain tissue). However, migration in highly confined environments was similar across the cell types. Modest differences were found in the stiffness of MDA-BR and MDA-BO cells plated on collagen I vs. fibronectin-coated surfaces. Lastly, MDA-BO cells were found to have larger focal adhesion area and density in comparison with the other two cell types. These results initiate a quantitative profile of mechanobiological phenotypes in TNBC, with future impacts aiming to help predict metastatic propensities to organ-specific sites in a clinical setting.
Collapse
Affiliation(s)
- Ariana Joy L. DeCastro
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Marina A. Pranda
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Kelsey M. Gray
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - John Merlo-Coyne
- Department of Biology, University of Maryland, College Park, MD, United States
| | - Nathaniel Girma
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Madelyn Hurwitz
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Yuji Zhang
- Department of Epidemiology and Public Health, University of Maryland, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, United States
| | - Kimberly M. Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, United States
- Biophysics Program, University of Maryland, College Park, MD, United States
- Center for Stem Cell Biology and Regenerative Medicine, University of Maryland, Baltimore, MD, United States
- *Correspondence: Kimberly M. Stroka,
| |
Collapse
|
17
|
Discovering the Triad between Nav1.5, Breast Cancer, and the Immune System: A Fundamental Review and Future Perspectives. Biomolecules 2022; 12:biom12020310. [PMID: 35204811 PMCID: PMC8869595 DOI: 10.3390/biom12020310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 02/12/2022] [Accepted: 02/13/2022] [Indexed: 02/05/2023] Open
Abstract
Nav1.5 is one of the nine voltage-gated sodium channel-alpha subunit (VGSC-α) family members. The Nav1.5 channel typically carries an inward sodium ion current that depolarises the membrane potential during the upstroke of the cardiac action potential. The neonatal isoform of Nav1.5, nNav1.5, is produced via VGSC-α alternative splicing. nNav1.5 is known to potentiate breast cancer metastasis. Despite their well-known biological functions, the immunological perspectives of these channels are poorly explored. The current review has attempted to summarise the triad between Nav1.5 (nNav1.5), breast cancer, and the immune system. To date, there is no such review available that encompasses these three components as most reviews focus on the molecular and pharmacological prospects of Nav1.5. This review is divided into three major subsections: (1) the review highlights the roles of Nav1.5 and nNav1.5 in potentiating the progression of breast cancer, (2) focuses on the general connection between breast cancer and the immune system, and finally (3) the review emphasises the involvements of Nav1.5 and nNav1.5 in the functionality of the immune system and the immunogenicity. Compared to the other subsections, section three is pretty unexploited; it would be interesting to study this subsection as it completes the triad.
Collapse
|
18
|
Chu YH, Liao WC, Ho YJ, Huang CH, Tseng TJ, Liu CH. Targeting Chondroitin Sulfate Reduces Invasiveness of Glioma Cells by Suppressing CD44 and Integrin β1 Expression. Cells 2021; 10:3594. [PMID: 34944101 PMCID: PMC8700349 DOI: 10.3390/cells10123594] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/14/2021] [Accepted: 12/17/2021] [Indexed: 12/18/2022] Open
Abstract
Chondroitin sulfate (CS) is a major component of the extracellular matrix found to be abnormally accumulated in several types of cancer tissues. Previous studies have indicated that CS synthases and modification enzymes are frequently elevated in human gliomas and are associated with poor prognosis. However, the underlying mechanisms of CS in cancer progression and approaches for interrupting its functions in cancer cells remain largely unexplored. Here, we have found that CS was significantly enriched surrounding the vasculature in a subset of glioma tissues, which was akin to the perivascular niche for cancer-initiating cells. Silencing or overexpression of the major CS synthase, chondroitin sulfate synthase 1 (CHSY1), significantly regulated the glioma cell invasive phenotypes and modulated integrin expression. Furthermore, we identified CD44 as a crucial chondroitin sulfate proteoglycan (CSPG) that was modified by CHSY1 on glioma cells, and the suppression of CS formation on CD44 by silencing the CHSY1-inhibited interaction between CD44 and integrin β1 on the adhesion complex. Moreover, we tested the CS-specific binding peptide, resulting in the suppression of glioma cell mobility in a fashion similar to that observed upon the silencing of CHSY1. In addition, the peptide demonstrated significant affinity to CD44, promoted CD44 degradation, and suppressed integrin β1 expression in glioma cells. Overall, this study proposes a potential regulatory loop between CS, CD44, and integrin β1 in glioma cells, and highlights the importance of CS in CD44 stability. Furthermore, the targeting of CS by specific binding peptides has potential as a novel therapeutic strategy for glioma.
Collapse
Affiliation(s)
- Yin-Hung Chu
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical University, Taichung 402306, Taiwan; (Y.-H.C.); (W.-C.L.); (C.-H.H.); (T.-J.T.)
| | - Wen-Chieh Liao
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical University, Taichung 402306, Taiwan; (Y.-H.C.); (W.-C.L.); (C.-H.H.); (T.-J.T.)
- Department of Medical Education, Chung Shan Medical University Hospital, Taichung 402306, Taiwan
| | - Ying-Jui Ho
- Department of Psychology, Chung Shan Medical University, Taichung 402306, Taiwan;
| | - Chih-Hsien Huang
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical University, Taichung 402306, Taiwan; (Y.-H.C.); (W.-C.L.); (C.-H.H.); (T.-J.T.)
| | - To-Jung Tseng
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical University, Taichung 402306, Taiwan; (Y.-H.C.); (W.-C.L.); (C.-H.H.); (T.-J.T.)
- Department of Medical Education, Chung Shan Medical University Hospital, Taichung 402306, Taiwan
| | - Chiung-Hui Liu
- Department of Anatomy, Faculty of Medicine, Chung Shan Medical University, Taichung 402306, Taiwan; (Y.-H.C.); (W.-C.L.); (C.-H.H.); (T.-J.T.)
- Department of Medical Education, Chung Shan Medical University Hospital, Taichung 402306, Taiwan
| |
Collapse
|
19
|
Deciphering the Molecular Machinery-Influence of sE-Cadherin on Tumorigenic Traits of Prostate Cancer Cells. BIOLOGY 2021; 10:biology10101007. [PMID: 34681106 PMCID: PMC8533516 DOI: 10.3390/biology10101007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 11/16/2022]
Abstract
Simple Summary Despite recent advances in the therapeutic management of metastasized prostate cancer, disease progression is still inevitable, with often fatal outcomes. Elucidating molecular mechanisms crucial to cancer development and progression is therefore necessary to find ways to interfere in metastatic processes and ultimately improve prognosis. Since soluble (s)E-cadherin is elevated in the serum of patients with prostate cancer, we investigated its influence on prostate cancer cell behavior in vitro. Exposure to sE-cadherin increased the systemic spread of the cells. Thus, targeting sE-cadherin might be a novel and innovative concept to treat advanced PCa. Abstract The serum level of soluble (s)E-cadherin is elevated in several malignancies, including prostate cancer (PCa). This study was designed to investigate the effects of sE-cadherin on the behavior of PCa cells in vitro, with the aim of identifying a potential therapeutic target. Growth as well as adhesive and motile behavior were evaluated in PC3, DU-145, and LNCaP cells. Flow cytometry was used to assess cell cycle phases and the surface expression of CD44 variants as well as α and β integrins. Confocal microscopy was utilized to visualize the distribution of CD44 variants within the cells. Western blot was applied to investigate expression of α3 and β1 integrins as well as cytoskeletal and adhesion proteins. Cell growth was significantly inhibited after exposure to 5 µg/mL sE-cadherin and was accompanied by a G0/G1-phase arrest. Adhesion of cells to collagen and fibronectin was mitigated, while motility was augmented. CD44v4, v5, and v7 expression was elevated while α3 and β1 integrins were attenuated. Blocking integrin α3 reduced cell growth and adhesion to collagen but increased motility. sE-cadherin therefore appears to foster invasive tumor cell behavior, and targeting it might serve as a novel and innovative concept to treat advanced PCa.
Collapse
|
20
|
Offeddu GS, Hajal C, Foley CR, Wan Z, Ibrahim L, Coughlin MF, Kamm RD. The cancer glycocalyx mediates intravascular adhesion and extravasation during metastatic dissemination. Commun Biol 2021; 4:255. [PMID: 33637851 PMCID: PMC7910477 DOI: 10.1038/s42003-021-01774-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 01/25/2021] [Indexed: 02/07/2023] Open
Abstract
The glycocalyx on tumor cells has been recently identified as an important driver for cancer progression, possibly providing critical opportunities for treatment. Metastasis, in particular, is often the limiting step in the survival to cancer, yet our understanding of how tumor cells escape the vascular system to initiate metastatic sites remains limited. Using an in vitro model of the human microvasculature, we assess here the importance of the tumor and vascular glycocalyces during tumor cell extravasation. Through selective manipulation of individual components of the glycocalyx, we reveal a mechanism whereby tumor cells prepare an adhesive vascular niche by depositing components of the glycocalyx along the endothelium. Accumulated hyaluronic acid shed by tumor cells subsequently mediates adhesion to the endothelium via the glycoprotein CD44. Trans-endothelial migration and invasion into the stroma occurs through binding of the isoform CD44v to components of the sub-endothelial extra-cellular matrix. Targeting of the hyaluronic acid-CD44 glycocalyx complex results in significant reduction in the extravasation of tumor cells. These studies provide evidence of tumor cells repurposing the glycocalyx to promote adhesive interactions leading to cancer progression. Such glycocalyx-mediated mechanisms may be therapeutically targeted to hinder metastasis and improve patient survival.
Collapse
Affiliation(s)
- Giovanni S. Offeddu
- grid.116068.80000 0001 2341 2786Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
| | - Cynthia Hajal
- grid.116068.80000 0001 2341 2786Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
| | - Colleen R. Foley
- grid.116068.80000 0001 2341 2786Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
| | - Zhengpeng Wan
- grid.116068.80000 0001 2341 2786Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
| | - Lina Ibrahim
- grid.116068.80000 0001 2341 2786Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
| | - Mark F. Coughlin
- grid.116068.80000 0001 2341 2786Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
| | - Roger D. Kamm
- grid.116068.80000 0001 2341 2786Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA USA ,grid.116068.80000 0001 2341 2786Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
| |
Collapse
|
21
|
Luo Y, Hu J, Liu Y, Li L, Li Y, Sun B, Kong R. Invadopodia: A potential target for pancreatic cancer therapy. Crit Rev Oncol Hematol 2021; 159:103236. [PMID: 33482351 DOI: 10.1016/j.critrevonc.2021.103236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 01/05/2021] [Accepted: 01/16/2021] [Indexed: 02/08/2023] Open
Abstract
Dissemination of cancer cells is an intricate multistep process that represents the most deadly aspect of cancer. Cancer cells form F-actin-rich protrusions known as invadopodia to invade surrounding tissues, blood vessels and lymphatics. A number of studies have demonstrated the significant roles of invadopodia in cancer. Therefore, the specific cells and molecules involved in invadopodia activity can provide as therapeutic targets. In this review, we included a thorough overview of studies in invadopodia and discussed their functions in cancer metastasis. We then presented the specific cells and molecules involved in invadopodia activity in pancreatic cancer and analyzed their suitability to be effective therapeutic targets. Currently, drugs targeting invadopodia and relevant clinical trials are negligible. Here, we highlighted the significance of potential drugs and discussed future obstacles in implementing clinical trials. This review presents a new perspective on invadopodia-induced pancreatic cancer metastasis and may prosper the development of targeted therapeutics against pancreatic cancer.
Collapse
Affiliation(s)
- Yan Luo
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jisheng Hu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yong Liu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Le Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yilong Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Rui Kong
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
22
|
Medrano-González PA, Rivera-Ramírez O, Montaño LF, Rendón-Huerta EP. Proteolytic Processing of CD44 and Its Implications in Cancer. Stem Cells Int 2021; 2021:6667735. [PMID: 33505471 PMCID: PMC7811561 DOI: 10.1155/2021/6667735] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/04/2020] [Accepted: 12/24/2020] [Indexed: 01/16/2023] Open
Abstract
CD44 is a transmembrane glycoprotein expressed in several healthy and tumor tissues. Modifications in its structure contribute differently to the activity of this molecule. One modification that has provoked interest is the consecutive cleavage of the CD44 extracellular ectodomain by enzymes that belong mainly to the family of metalloproteases. This process releases biologically active substrates, via alternative splice forms of CD44, that generate CD44v3 or v6 isoforms which participate in the transcriptional regulation of genes and proteins associated to signaling pathways involved in the development of cancer. These include the protooncogene tyrosine-protein kinase Src (c-Src)/signal transducer and activator of transcription 3 (STAT3), the epithelial growth factor receptor, the estrogen receptor, Wnt/βcatenin, or Hippo signaling pathways all of which are associated to cell proliferation, differentiation, or cancer progression. Whereas CD44 still remains as a very useful prognostic cell marker in different pathologies, the main topic is that the generation of CD44 intracellular fragments assists the regulation of transcriptional proteins involved in the cell cycle, cell metabolism, and most importantly, the regulation of some stem cell-associated markers.
Collapse
Affiliation(s)
- Priscila Anhel Medrano-González
- Lab. Inmunobiología, Depto. Biología Celular y Tisular, Facultad de Medicina, UNAM, Mexico, Mexico
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Edif. D, 1 piso, Circuito de Posgrados, Ciudad Universitaria, Coyoacán, 04510 Mexico, Mexico
| | - Osmar Rivera-Ramírez
- Lab. Inmunobiología, Depto. Biología Celular y Tisular, Facultad de Medicina, UNAM, Mexico, Mexico
| | - Luis Felipe Montaño
- Lab. Inmunobiología, Depto. Biología Celular y Tisular, Facultad de Medicina, UNAM, Mexico, Mexico
| | - Erika P. Rendón-Huerta
- Lab. Inmunobiología, Depto. Biología Celular y Tisular, Facultad de Medicina, UNAM, Mexico, Mexico
| |
Collapse
|
23
|
Hou J, Yan D, Liu Y, Huang P, Cui H. The Roles of Integrin α5β1 in Human Cancer. Onco Targets Ther 2020; 13:13329-13344. [PMID: 33408483 PMCID: PMC7781020 DOI: 10.2147/ott.s273803] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/21/2020] [Indexed: 12/19/2022] Open
Abstract
Cell adhesion to the extracellular matrix has important roles in tissue integrity and human health. Integrins are heterodimeric cell surface receptors that are composed by two non-covalently linked alpha and beta subunits that mainly participate in the interaction of cell-cell adhesion and cell-extracellular matrix and regulate cell motility, adhesion, differentiation, migration, proliferation, etc. In mammals, there have been eighteen α subunits and 8 β subunits and so far 24 distinct types of αβ integrin heterodimers have been identified in humans. Integrin α5β1, also known as the fibronectin receptor, is a heterodimer with α5 and β1 subunits and has emerged as an essential mediator in many human carcinomas. Integrin α5β1 alteration is closely linked to the progression of several types of human cancers, including cell proliferation, angiogenesis, tumor metastasis, and cancerogenesis. In this review, we will introduce the functions of integrin α5β1 in cancer progression and also explore its regulatory mechanisms. Additionally, the potential clinical applications as a target for cancer imaging and therapy are discussed. Collectively, the information reviewed here may increase the understanding of integrin α5β1 as a potential therapeutic target for cancer.
Collapse
Affiliation(s)
- Jianbing Hou
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing 400716, People's Republic of China.,Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, People's Republic of China.,Chongqing Engineering and Technology Research Centre for Silk Biomaterials and Regenerative Medicine, Chongqing 400716, People's Republic of China
| | - Du Yan
- Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing 400716, People's Republic of China
| | - Yudong Liu
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing 400716, People's Republic of China.,Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, People's Republic of China.,Chongqing Engineering and Technology Research Centre for Silk Biomaterials and Regenerative Medicine, Chongqing 400716, People's Republic of China
| | - Pan Huang
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing 400716, People's Republic of China.,Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, People's Republic of China.,Chongqing Engineering and Technology Research Centre for Silk Biomaterials and Regenerative Medicine, Chongqing 400716, People's Republic of China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing 400716, People's Republic of China.,Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, People's Republic of China.,Chongqing Engineering and Technology Research Centre for Silk Biomaterials and Regenerative Medicine, Chongqing 400716, People's Republic of China
| |
Collapse
|
24
|
Rutz J, Thaler S, Maxeiner S, Chun FKH, Blaheta RA. Sulforaphane Reduces Prostate Cancer Cell Growth and Proliferation In Vitro by Modulating the Cdk-Cyclin Axis and Expression of the CD44 Variants 4, 5, and 7. Int J Mol Sci 2020; 21:ijms21228724. [PMID: 33218199 PMCID: PMC7699211 DOI: 10.3390/ijms21228724] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer patients whose tumors develop resistance to conventional treatment often turn to natural, plant-derived products, one of which is sulforaphane (SFN). This study was designed to determine whether anti-tumor properties of SFN, identified in other tumor entities, are also evident in cultivated DU145 and PC3 prostate cancer cells. The cells were incubated with SFN (1–20 µM) and tumor cell growth and proliferative activity were evaluated. Having found a considerable anti-growth, anti-proliferative, and anti-clonogenic influence of SFN on both prostate cancer cell lines, further investigation into possible mechanisms of action were performed by evaluating the cell cycle phases and cell-cycle-regulating proteins. SFN induced a cell cycle arrest at the S- and G2/M-phase in both DU145 and PC3 cells. Elevation of histone H3 and H4 acetylation was also evident in both cell lines following SFN exposure. However, alterations occurring in the Cdk-cyclin axis, modification of the p19 and p27 proteins and changes in CD44v4, v5, and v7 expression because of SFN exposure differed in the two cell lines. SFN, therefore, does exert anti-tumor properties on these two prostate cancer cell lines by histone acetylation and altering the intracellular signaling cascade, but not through the same molecular mechanisms.
Collapse
|
25
|
Breast Cancer-Derived Microparticles Reduce Cancer Cell Adhesion, an Effect Augmented by Chemotherapy. Cells 2020; 9:cells9102269. [PMID: 33050539 PMCID: PMC7650796 DOI: 10.3390/cells9102269] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/08/2020] [Accepted: 10/10/2020] [Indexed: 12/24/2022] Open
Abstract
Tumor cell heterogeneity is primarily dictated by mutational changes, sometimes leading to clones that undergo a metastatic switch. However, little is known about tumor heterogeneity following chemotherapy perturbation. Here we studied the possible involvement of tumor-derived extracellular vesicles, often referred to as tumor-derived microparticles (TMPs), as mediators of the metastatic switch in the tumor microenvironment by hindering cell adhesion properties. Specifically, we show that highly metastatic or chemotherapy-treated breast cancer cells shed an increased number of TMPs compared to their respective controls. We found that these TMPs substantially reduce cell adhesion and disrupt actin filament structure, therefore increasing their biomechanical force pace, further implicating tumor cell dissemination as part of the metastatic cascade. Our results demonstrate that these pro-metastatic effects are mediated in part by CD44 which is highly expressed in TMPs obtained from highly metastatic cells or cells exposed to chemotherapy when compared to cells with low metastatic potential. Consequently, when we inhibited CD44 expression on TMPs by a pharmacological or a genetic approach, increased tumor cell adhesion and re-organized actin filament structure were observed. We also demonstrated that breast cancer patients treated with paclitaxel chemotherapy exhibited increased CD44-expressing TMPs. Overall, our study provides further insights into the role of TMPs in promoting metastasis, an effect which is augmented when tumor cells are exposed to chemotherapy.
Collapse
|
26
|
Multilayer platform to model the bioactivity of hyaluronic acid in gastric cancer. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 119:111616. [PMID: 33321659 DOI: 10.1016/j.msec.2020.111616] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 09/10/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023]
Abstract
Hyaluronic acid (HA) has a key role in cancer progression. The HA's molecular weight (Mw) is altered in this pathological state: increased concentration of shorter fragments due to the overexpressed hyaluronidases and ROS. Aiming to mimic this microenvironment, we developed a Layer-by-Layer (LbL) platform presenting HA of different Mws, namely 6.4, 752 and 1500 kDa, to study the influence of HA Mw on the formation of focal adhesion sites (FAs), and the involvement of paxillin and CD44 in this process. High paxillin expression and formation of FAs, via CD44, is observed for MKN45 cells seeded on LbLs presenting HA 6.4 kDa, with the activation of the ERK1/2 pathway, responsible for cell motility and tumour progression. In contrast, activation of p38 pathway, usually related with cancer latency, is observed for cells seeded on LbLs with high Mw HA, i.e. 1500 kDa. Overall, we demonstrate the suitability of the developed platform to study cancer invasiveness.
Collapse
|
27
|
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: 66] [Impact Index Per Article: 13.2] [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
|
28
|
Cruz da Silva E, Dontenwill M, Choulier L, Lehmann M. Role of Integrins in Resistance to Therapies Targeting Growth Factor Receptors in Cancer. Cancers (Basel) 2019; 11:cancers11050692. [PMID: 31109009 PMCID: PMC6562376 DOI: 10.3390/cancers11050692] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/13/2019] [Accepted: 05/14/2019] [Indexed: 02/07/2023] Open
Abstract
Integrins contribute to cancer progression and aggressiveness by activating intracellular signal transduction pathways and transducing mechanical tension forces. Remarkably, these adhesion receptors share common signaling networks with receptor tyrosine kinases (RTKs) and support their oncogenic activity, thereby promoting cancer cell proliferation, survival and invasion. During the last decade, preclinical studies have revealed that integrins play an important role in resistance to therapies targeting RTKs and their downstream pathways. A remarkable feature of integrins is their wide-ranging interconnection with RTKs, which helps cancer cells to adapt and better survive therapeutic treatments. In this context, we should consider not only the integrins expressed in cancer cells but also those expressed in stromal cells, since these can mechanically increase the rigidity of the tumor microenvironment and confer resistance to treatment. This review presents some of these mechanisms and outlines new treatment options for improving the efficacy of therapies targeting RTK signaling.
Collapse
Affiliation(s)
- Elisabete Cruz da Silva
- UMR 7021 CNRS, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Université de Strasbourg, Faculté de Pharmacie, 67401 Illkirch, France.
| | - Monique Dontenwill
- UMR 7021 CNRS, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Université de Strasbourg, Faculté de Pharmacie, 67401 Illkirch, France.
| | - Laurence Choulier
- UMR 7021 CNRS, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Université de Strasbourg, Faculté de Pharmacie, 67401 Illkirch, France.
| | - Maxime Lehmann
- UMR 7021 CNRS, Laboratoire de Bioimagerie et Pathologies, Tumoral Signaling and Therapeutic Targets, Université de Strasbourg, Faculté de Pharmacie, 67401 Illkirch, France.
| |
Collapse
|
29
|
Zhang X, Xing XX, Cui JF. Invadopodia formation: An important step in matrix stiffness-regulated tumor invasion and metastasis. Shijie Huaren Xiaohua Zazhi 2019; 27:589-597. [DOI: 10.11569/wcjd.v27.i9.589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Highly motile and invasive abilities are symbolic features of metastatic tumor cells. Being a critical molecular event for maintaining the highly migratory and invasive capabilities of tumor cells, invadopodia formation undoubtedly determines the progression of tumor invasion and metastasis. Growing numbers of studies suggest that increased matrix stiffness, as a notable property of physical mechanics in solid tumors, participates in the regulation of tumor invasion and metastasis via different molecular mechanisms. However, to date the relevant mechanisms of matrix stiffness-induced invadopodia formation and activity in tumor cells remain largely unclear. This paper is to make a review on the structure and function of invadopodia, the stages and inductive factors of invadopodia formation, the regulatory mechanisms of matrix stiffness-induced invadopodia formation and so on, with an aim to reveal the important roles of invadopodia in matrix stiffness-regulated tumor invasion and metastasis.
Collapse
Affiliation(s)
- Xi Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiao-Xia Xing
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jie-Feng Cui
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
30
|
Xu Z, Isaji T, Fukuda T, Wang Y, Gu J. O-GlcNAcylation regulates integrin-mediated cell adhesion and migration via formation of focal adhesion complexes. J Biol Chem 2018; 294:3117-3124. [PMID: 30587575 DOI: 10.1074/jbc.ra118.005923] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/02/2018] [Indexed: 01/08/2023] Open
Abstract
O-GlcNAcylation is a post-translational modification of a protein serine or threonine residue catalyzed by O-GlcNAc transferase (OGT) in the nucleus and cytoplasm. O-GlcNAcylation plays important roles in the cellular signaling that affect the different biological functions of cells, depending upon cell type. However, whether or not O-GlcNAcylation regulates cell adhesion and migration remains unclear. Here, we used the doxycycline-inducible short hairpin RNA (shRNA) system to establish an OGT knockdown (KD) HeLa cell line and found that O-GlcNAcylation is a key regulator for cell adhesion, migration, and focal adhesion (FA) complex formation. The expression levels of OGT and O-GlcNAcylation were remarkably suppressed 24 h after induction of doxycycline. Knockdown of OGT significantly promoted cell adhesion, but it suppressed the cell migration on fibronectin. The immunostaining with paxillin, a marker for FA plaque, clearly showed that the number of FAs was increased in the KD cells compared with that in the control cells. The O-GlcNAcylation levels of paxillin, talin, and focal adhesion kinase were down-regulated in KD cells. Interestingly, the complex formation between integrin β1, focal adhesion kinase, paxillin, and talin was greatly increased in KD cells. Consistently, levels of active integrin β1 were significantly enhanced in KD cells, whereas they were decreased in cells overexpressing OGT. The data suggest a novel regulatory mechanism for O-GlcNAcylation during FA complex formation, which thereby affects integrin activation and integrin-mediated functions such as cell adhesion and migration.
Collapse
Affiliation(s)
- Zhiwei Xu
- From the Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai Miyagi 981-8558, Japan and
| | - Tomoya Isaji
- From the Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai Miyagi 981-8558, Japan and
| | - Tomohiko Fukuda
- From the Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai Miyagi 981-8558, Japan and
| | - Yuqin Wang
- the Department of Pharmacology, Pharmacy College, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Jianguo Gu
- From the Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai Miyagi 981-8558, Japan and
| |
Collapse
|
31
|
The CD44 variant induces K562 cell acquired with resistance to adriamycin via NF-κB/Snail/Bcl-2 pathway. Med Hypotheses 2018; 121:142-148. [DOI: 10.1016/j.mehy.2018.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 08/09/2018] [Accepted: 09/05/2018] [Indexed: 01/28/2023]
|
32
|
Ouhtit A, Rizeq B, Saleh HA, Rahman MM, Zayed H. Novel CD44-downstream signaling pathways mediating breast tumor invasion. Int J Biol Sci 2018; 14:1782-1790. [PMID: 30443182 PMCID: PMC6231220 DOI: 10.7150/ijbs.23586] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 02/04/2018] [Indexed: 01/06/2023] Open
Abstract
CD44, also known as homing cell adhesion molecule is a multi-structural cell molecule involved in cell-cell and cell-extracellular matrix communications. CD44 regulates a number of central signaling pathways, including PI3K/AKT, Rho GTPases and the Ras-MAPK pathways, but also acts as a growth/arrest sensor, and inhibitor of angiogenesis and invasion, in response to signals from the microenvironment. The function of CD44 has been very controversial since it acts as both, a suppressor and a promoter of tumor growth and progression. To address this discrepancy, we have previously established CD44-inducible system both in vitro and in vivo. Next, using microarray analysis, we have identified and validated Survivin, Cortactin and TGF-β2 as novel CD44-downstream target genes, and characterized their signaling pathways underpinning CD44-promoted breast cancer (BC) cell invasion. This report aims to update the literature by adding and discussing the impact of these novel three signaling pathways to better understand the CD44-signaling pathways involved in BC tumor cell invasion.
Collapse
Affiliation(s)
- Allal Ouhtit
- Department of Biological and Environmental Sciences, College of Arts & Sciences, Qatar University, Doha, Qatar
| | - Balsam Rizeq
- Department of Biological and Environmental Sciences, College of Arts & Sciences, Qatar University, Doha, Qatar.,Biomedical Research Center, Qatar University, Doha, Qatar
| | - Haissam Abou Saleh
- Department of Biological and Environmental Sciences, College of Arts & Sciences, Qatar University, Doha, Qatar
| | - Md Mizanur Rahman
- Department of Biological and Environmental Sciences, College of Arts & Sciences, Qatar University, Doha, Qatar
| | - Hatem Zayed
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar
| |
Collapse
|
33
|
Li X, Wang S, Yang S, Ying J, Yu H, Yang C, Liu Y, Wang Y, Cheng S, Xiao J, Guo H, Jiang Z, Wang Z. Circadian locomotor output cycles kaput affects the proliferation and migration of breast cancer cells by regulating the expression of E-cadherin via IQ motif containing GTPase activating protein 1. Oncol Lett 2018; 15:7097-7103. [PMID: 29731875 PMCID: PMC5920822 DOI: 10.3892/ol.2018.8226] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 01/19/2018] [Indexed: 12/29/2022] Open
Abstract
The circadian rhythm regulates numerous physiological activities, including sleep and wakefulness, behavior, immunity and metabolism. Previous studies have demonstrated that circadian rhythm disorder is associated with the occurrence of tumors. Responsible for regulating a number of functions, the Circadian locomotor output cycles kaput (Clock) gene is one of the core regulatory genes of circadian rhythm. The Clock gene has also been implicated in the occurrence and development of tumors in previously studies. The present study evaluated the role of the Clock gene in the proliferation and migration of mouse breast cancer 4T1 cells, and investigated its possible regulatory pathways and mechanisms. It was reported that downregulation of Clock facilitated the proliferation and migration of breast cancer cells. Further investigation revealed the involvement of IQ motif containing GTPase activating protein 1 (IQGAP1) protein expression in the Clock regulatory pathway, further influencing the expression of E-cadherin, a known proprietor of tumor cell migration and invasion. To the best of our knowledge, the present study is the first to report that Clock, acting through the regulation of the scaffolding protein IQGAP1, regulates the downstream expression of E-cadherin, thereby affecting tumor cell structure and motility. These results confirmed the role of Clock in breast cancer tumor etiology and provide insight regarding the molecular avenues of its regulatory nature, which may translate beyond breast cancer into other known functions of the gene.
Collapse
Affiliation(s)
- Xiaoxue Li
- Health Ministry Key Laboratory of Chronobiology, College of Basic Medicine and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Siyang Wang
- Department of Life Sciences, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Shuhong Yang
- Health Ministry Key Laboratory of Chronobiology, College of Basic Medicine and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Junjie Ying
- Health Ministry Key Laboratory of Chronobiology, College of Basic Medicine and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hang Yu
- Health Ministry Key Laboratory of Chronobiology, College of Basic Medicine and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Chunlei Yang
- Department of Life Sciences, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yanyou Liu
- Health Ministry Key Laboratory of Chronobiology, College of Basic Medicine and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yuhui Wang
- Health Ministry Key Laboratory of Chronobiology, College of Basic Medicine and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Shuting Cheng
- Health Ministry Key Laboratory of Chronobiology, College of Basic Medicine and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jing Xiao
- Health Ministry Key Laboratory of Chronobiology, College of Basic Medicine and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Huiling Guo
- Health Ministry Key Laboratory of Chronobiology, College of Basic Medicine and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Zhou Jiang
- Health Ministry Key Laboratory of Chronobiology, College of Basic Medicine and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Zhengrong Wang
- Health Ministry Key Laboratory of Chronobiology, College of Basic Medicine and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
34
|
Weitzenfeld P, Meshel T, Ben-Baruch A. Microenvironmental networks promote tumor heterogeneity and enrich for metastatic cancer stem-like cells in Luminal-A breast tumor cells. Oncotarget 2018; 7:81123-81143. [PMID: 27835603 PMCID: PMC5348381 DOI: 10.18632/oncotarget.13213] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 11/02/2016] [Indexed: 12/12/2022] Open
Abstract
The roles of the tumor microenvironment (TME) in generating intra-tumoral diversity within each specific breast cancer subtype are far from being fully elucidated. In this study, we exposed Luminal-A breast cancer cells in culture to combined “TME Stimulation”, representing three typical arms of the breast TME: hormonal (estrogen), inflammatory (tumor necrosis factor α) and growth-promoting (epidermal growth factor). In addition to enriching the tumor cell population with CD44+/β1+ cells (as we previously published), TME Stimulation selected for CD44+/CD24low/− stem-like cells, that were further enriched by doxorubicin treatment and demonstrated high plasticity in vitro and in vivo. Knock-down experiments revealed that CD44 and Zeb1 regulated CD24 and β1 expression and controlled differently cell spreading and formation of cellular protrusions. TME-enriched CD44+/CD24low/− stem-like cells promoted dissemination to bones and lymph nodes, whereas CD44+/β1+ cells had a low metastatic potential. Mixed co-injections of TME-enriched CD44+/CD24low/− and CD44+/β1+ sub-populations generated metastases populated mostly by CD44+/CD24low/−-derived cells. Thus, combined activities of several TME factors select for CD44+/CD24low/− stem-like cells that dictate the metastatic phenotype of Luminal-A breast tumor cells, suggesting that therapeutic modalities targeting the TME could be introduced as a potential strategy of inhibiting the detrimental stem-like sub-population in this disease subtype.
Collapse
Affiliation(s)
- Polina Weitzenfeld
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Tsipi Meshel
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Adit Ben-Baruch
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
35
|
Wang Z, von Au A, Schnölzer M, Hackert T, Zöller M. CD44v6-competent tumor exosomes promote motility, invasion and cancer-initiating cell marker expression in pancreatic and colorectal cancer cells. Oncotarget 2018; 7:55409-55436. [PMID: 27419629 PMCID: PMC5342426 DOI: 10.18632/oncotarget.10580] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Accepted: 07/01/2016] [Indexed: 12/21/2022] Open
Abstract
Cancer-initiating cells (CIC) account for metastatic spread, which may rely mostly on CIC exosomes (TEX) that affect host cells and can transfer CIC features into Non-CIC. The CIC marker CD44 variant isoform v6 (CD44v6) being known for metastasis-promotion, we elaborated in cells its contribution to migration and invasion and in TEX the tranfer of migratory and invasive capacity to Non-CIC, using a CD44v6 knockdown (CD44v6kd) as Non-CIC model.A CD44v6kd in human pancreatic and colorectal cancer (PaCa, CoCa) lines led to loss of CIC characteristics including downregulation of additional CIC markers, particularly Tspan8. This aggravated the loss of CD44v6-promoted motility and invasion. Loss of motility relies on the distorted cooperation of CD44v6 and Tspan8 with associated integrins and loss of invasiveness on reduced protease expression. These deficits, transferred into TEX, severely altered the CD44v6kd-TEX composition. As a consequence, unlike the CIC-TEX, CD44v6kd TEX were not taken up by CD44v6kd cells and CIC. The uptake of CIC-TEX was accompanied by partial correction of CIC marker and protease expression in CD44v6kd cells, which regained migratory, invasive and metastatic competence. CIC-TEX also fostered angiogenesis and expansion of myeloid cells, likely due to a direct impact of CIC-TEX on the host, which could be supported by reprogrammed CD44v6kd cells.Taken together, the striking loss of tumor progression by a CD44v6kd relies on the capacity of CD44v6 to cooperate with associating integrins and proteases and its promotion of additional CIC marker expression. The defects by a CD44v6kd are efficiently corrected upon CIC-TEX uptake.
Collapse
Affiliation(s)
- Zhe Wang
- Tumor Cell Biology, University Hospital of Surgery, Heidelberg, Germany
| | - Anja von Au
- Tumor Cell Biology, University Hospital of Surgery, Heidelberg, Germany
| | - Martina Schnölzer
- Proteome Analysis Department, German Cancer Research Center, Heidelberg, Germany
| | - Thilo Hackert
- Section Pancreas Research, University Hospital of Surgery, Heidelberg, Germany
| | - Margot Zöller
- Tumor Cell Biology, University Hospital of Surgery, Heidelberg, Germany
| |
Collapse
|
36
|
Huang GX, Qi MF, Li XL, Tang F, Zhu L. Involvement of upregulation of fibronectin in the pro‑adhesive and pro‑survival effects of glucocorticoid on melanoma cells. Mol Med Rep 2017; 17:3380-3387. [PMID: 29257300 DOI: 10.3892/mmr.2017.8269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Accepted: 12/05/2017] [Indexed: 11/06/2022] Open
Abstract
Glucocorticoids (GCs) are important stress hormones, which are used as a concomitant medication during malignant tumor chemotherapy. Clinical and preclinical studies have linked GCs to melanoma growth and progression. However, the effects and mechanism of action of GCs on the adhesion and survival of melanoma cells are still unknown. In the present study the effect of dexamethasone (Dex), a synthetic GC, on fibronectin (FN) expression and its roles in regulating the adhesion and survival of melanoma cells were investigated. It was revealed that Dex significantly increased the levels of intracellular and secreted FN in melanoma cell lines by increasing glucocorticoid receptor‑mediated FN protein stability. Additionally, it was demonstrated that Dex (100 nM) significantly promoted the adhesion and survival of melanoma cells. Silencing FN expression abrogated the pro‑adhesive and pro‑survival effects of Dex in melanoma cells. Extracellular FN significantly enhanced melanoma cell adhesion and survival in the presence of cisplatin, whereas partially blocking extracellular FN signaling with a CD44 antibody significantly reduced FN‑enhanced adhesion and survival. This indicated that the upregulation of FN contributed to the pro‑survival effect of Dex by enhancing cell adhesion. It was also observed that activation of the PI3K/AKT signaling pathway by extracellular FN was involved in the FN‑mediated increase in melanoma cell survival. These findings increase understanding of the possible mechanisms by which GCs regulate melanoma cell adhesion and survival.
Collapse
Affiliation(s)
- Gao-Xiang Huang
- Department of Pathology, No. 181 Hospital of People's Liberation Army, Guilin, Guangxi 541002, P.R. China
| | - Min-Fang Qi
- Department of Pathology, No. 181 Hospital of People's Liberation Army, Guilin, Guangxi 541002, P.R. China
| | - Xiao-Long Li
- Department of Cardiology, Navy General Hospital, Beijing 100048, P.R. China
| | - Fang Tang
- Department of Pathology, No. 181 Hospital of People's Liberation Army, Guilin, Guangxi 541002, P.R. China
| | - Lei Zhu
- Department of Orthopedic Trauma Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| |
Collapse
|
37
|
Mayr L, Pirker C, Lötsch D, Van Schoonhoven S, Windhager R, Englinger B, Berger W, Kubista B. CD44 drives aggressiveness and chemoresistance of a metastatic human osteosarcoma xenograft model. Oncotarget 2017; 8:114095-114108. [PMID: 29371972 PMCID: PMC5768389 DOI: 10.18632/oncotarget.23125] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 11/28/2017] [Indexed: 12/15/2022] Open
Abstract
Background Osteosarcoma is the most common primary malignant bone tumor with a 5 year survival rate of up to 70%. However, patients with metastatic disease have still a very poor prognosis. Osteosarcoma metastasis models are essential to develop novel treatment strategies for advanced disease. Methods Based on a serial transplantation approach, we have established a U-2 OS osteosarcoma xenograft model with increased metastatic potential and compared it to other metastatic osteosarcoma models from international sources. Subclones with differing invasive potential were compared for genomic gains and losses as well as gene expression changes by several bioinformatic approaches. Based on the acquired results, the effects of a shRNA-mediated CD44 mRNA knockdown on migration, invasion and chemosensitivity were evaluated. Results The CD44 gene was part of an amplified region at chromosome 11p found in both U-2 OS subclones with enhanced metastatic potential but not in parental U-2 OS cells, corresponding with distinct CD44 overexpression. Accordingly, shRNA-mediated CD44 knockdown significantly attenuated osteosarcoma cell migration, invasion, and viability especially in the metastatic subclones of U-2 OS and Saos-2 cells. Metastatic subclones generally were hypersensitive against the integrin inhibitor cilengitide paralleled by alterations in integrin expression pattern following CD44 knock-down. Additionally, attenuation of CD44 expression sensitized these cell models against osteosarcoma chemotherapy with doxorubicin but not methotrexate and cisplatin. Conclusions The osteosarcoma xenograft models with increased metastatic potential developed in this study can be useful for identification of mechanisms driving metastasis and resistance towards clinically used and novel therapeutic regimens.
Collapse
Affiliation(s)
- Lisa Mayr
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University Vienna, 1090 Vienna, Austria
| | - Christine Pirker
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University Vienna, 1090 Vienna, Austria
| | - Daniela Lötsch
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University Vienna, 1090 Vienna, Austria
| | - Sushilla Van Schoonhoven
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University Vienna, 1090 Vienna, Austria
| | - Reinhard Windhager
- Department of Orthopaedics, Medical University Vienna, 1090 Vienna, Austria
| | - Bernhard Englinger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University Vienna, 1090 Vienna, Austria
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University Vienna, 1090 Vienna, Austria
| | - Bernd Kubista
- Department of Orthopaedics, Medical University Vienna, 1090 Vienna, Austria
| |
Collapse
|
38
|
Breuksch I, Prosinger F, Baehr F, Engelhardt FP, Bauer HK, Thüroff JW, Heimes AS, Hasenburg A, Prawitt D, Brenner W. Integrin α5 triggers the metastatic potential in renal cell carcinoma. Oncotarget 2017; 8:107530-107542. [PMID: 29296184 PMCID: PMC5746086 DOI: 10.18632/oncotarget.22501] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 10/28/2017] [Indexed: 12/24/2022] Open
Abstract
The therapy of advanced renal cell carcinoma (RCC) is still a major challenge. To intervene therapeutically a deeper comprehension of the particular steps of metastasis is necessary. In this context membrane bound receptors like integrins play a decisive role. We analyzed the integrin α5 expression in 141 clear cell RCC patients by Western blot. Patients with RCC expressed a significant higher level of integrin α5 in tumor than in normal tissue. The integrin α5 expression correlated with tumor grade, the development of distant metastases within five years after tumor nephrectomy and reduced survival. The RCC cell lines Caki-1 and CCF-RC1, which highly express integrin α5, were treated with fibronectin in combination with or without an inhibiting anti-integrin α5 antibody. Afterwards the migration, adhesion, viability and prominent signaling molecules were analyzed. Both cell lines showed a significant reduced migration potential as well as a decreased adhesion potential to fibronectin after treatment with an integrin α5 blocking antibody. A contribution of the AKT and ERK1/2 signaling pathways could be demonstrated. The results indicate integrin α5 as a potent marker to discriminate patients’ tumor prognosis. Consequently the integrin subunit α5 can be considered as a target for individual therapy of advanced RCC.
Collapse
Affiliation(s)
- Ines Breuksch
- Department of Gynecology, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany.,Department of Urology, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Franz Prosinger
- Department of Gynecology, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Fabian Baehr
- Department of Urology, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Franz-Peter Engelhardt
- Department of Urology, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Heide-Katharina Bauer
- Department of Gynecology, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Joachim W Thüroff
- Department of Urology, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Anne-Sophie Heimes
- Department of Gynecology, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Annette Hasenburg
- Department of Gynecology, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Dirk Prawitt
- Department of Pediatrics, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Walburgis Brenner
- Department of Gynecology, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany.,Department of Urology, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| |
Collapse
|
39
|
Malric L, Monferran S, Gilhodes J, Boyrie S, Dahan P, Skuli N, Sesen J, Filleron T, Kowalski-Chauvel A, Cohen-Jonathan Moyal E, Toulas C, Lemarié A. Interest of integrins targeting in glioblastoma according to tumor heterogeneity and cancer stem cell paradigm: an update. Oncotarget 2017; 8:86947-86968. [PMID: 29156849 PMCID: PMC5689739 DOI: 10.18632/oncotarget.20372] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 07/23/2017] [Indexed: 12/22/2022] Open
Abstract
Glioblastomas are malignant brain tumors with dismal prognosis despite standard treatment with surgery and radio/chemotherapy. These tumors are defined by an important cellular heterogeneity and notably contain a particular subpopulation of Glioblastoma-initiating cells, which recapitulate the heterogeneity of the original Glioblastoma. In order to classify these heterogeneous tumors, genomic profiling has also been undertaken to classify these heterogeneous tumors into several subtypes. Current research focuses on developing therapies, which could take into account this cellular and genomic heterogeneity. Among these targets, integrins are the subject of numerous studies since these extracellular matrix transmembrane receptors notably controls tumor invasion and progression. Moreover, some of these integrins are considered as membrane markers for the Glioblastoma-initiating cells subpopulation. We reviewed here integrin expression according to glioblastoma molecular subtypes and cell heterogeneity. We discussed their roles in glioblastoma invasion, angiogenesis, therapeutic resistance, stemness and microenvironment modulations, and provide an overview of clinical trials investigating integrins in glioblastomas. This review highlights that specific integrins could be identified as selective glioblastoma cells markers and that their targeting represents new diagnostic and/or therapeutic strategies.
Collapse
Affiliation(s)
- Laure Malric
- INSERM U1037, Center for Cancer Research of Toulouse, Toulouse, France
| | - Sylvie Monferran
- INSERM U1037, Center for Cancer Research of Toulouse, Toulouse, France.,Faculty of Pharmaceutical Sciences, University of Toulouse III Paul Sabatier, Toulouse, France
| | - Julia Gilhodes
- Department of Biostatistics, IUCT-Oncopole, Toulouse, France
| | - Sabrina Boyrie
- INSERM U1037, Center for Cancer Research of Toulouse, Toulouse, France
| | - Perrine Dahan
- INSERM U1037, Center for Cancer Research of Toulouse, Toulouse, France
| | - Nicolas Skuli
- INSERM U1037, Center for Cancer Research of Toulouse, Toulouse, France.,Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Julie Sesen
- INSERM U1037, Center for Cancer Research of Toulouse, Toulouse, France
| | - Thomas Filleron
- Department of Biostatistics, IUCT-Oncopole, Toulouse, France
| | | | - Elizabeth Cohen-Jonathan Moyal
- INSERM U1037, Center for Cancer Research of Toulouse, Toulouse, France.,Department of Radiotherapy, IUCT-Oncopole, Toulouse, France
| | - Christine Toulas
- INSERM U1037, Center for Cancer Research of Toulouse, Toulouse, France.,Laboratory of Oncogenetic, IUCT-Oncopole, Toulouse, France
| | - Anthony Lemarié
- INSERM U1037, Center for Cancer Research of Toulouse, Toulouse, France.,Faculty of Pharmaceutical Sciences, University of Toulouse III Paul Sabatier, Toulouse, France
| |
Collapse
|
40
|
Tumor Cell Invadopodia: Invasive Protrusions that Orchestrate Metastasis. Trends Cell Biol 2017; 27:595-607. [PMID: 28412099 DOI: 10.1016/j.tcb.2017.03.003] [Citation(s) in RCA: 256] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/09/2017] [Accepted: 03/13/2017] [Indexed: 12/26/2022]
Abstract
Invadopodia are a subset of invadosomes that are implicated in the integration of signals from the tumor microenvironment to support tumor cell invasion and dissemination. Recent progress has begun to define how tumor cells regulate the plasticity necessary for invadopodia to assemble and function efficiently in the different microenvironments encountered during dissemination in vivo. Exquisite mapping by many laboratories of the pathways involved in integrating diverse invadopodium initiation signals, from growth factors, to extracellular matrix (ECM) and cell-cell contact in the tumor microenvironment, has led to insight into the molecular basis of this plasticity. Here, we integrate this new information to discuss how the invadopodium is an important conductor that orchestrates tumor cell dissemination during metastasis.
Collapse
|
41
|
Zhu X, Zhou Y, Xu Q, Wu J. Traditional Chinese medicine Jianpi Bushen therapy suppresses the onset of pre-metastatic niche in a murine model of spontaneous lung metastasis. Biomed Pharmacother 2017; 86:434-440. [DOI: 10.1016/j.biopha.2016.12.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 11/18/2016] [Accepted: 12/05/2016] [Indexed: 11/25/2022] Open
|
42
|
Xu H, Wu K, Tian Y, Liu Q, Han N, Yuan X, Zhang L, Wu GS, Wu K. CD44 correlates with clinicopathological characteristics and is upregulated by EGFR in breast cancer. Int J Oncol 2016; 49:1343-50. [PMID: 27499099 PMCID: PMC5021250 DOI: 10.3892/ijo.2016.3639] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 07/15/2016] [Indexed: 12/15/2022] Open
Abstract
Cluster of differentiation 44 (CD44), a well-known transmembrane glycoprotein, serves as a promoting factor in the carcinogenesis and progression of a variety of neoplasms. Previous studies have demonstrated that aberrant expression of CD44 was associated with the initiation, invasion, metastasis, and therapy-resistance of breast cancer, but whether there was any association between CD44 and pathological characteristics of breast cancer or epidermal growth factor receptor (EGFR) has not been clearly elucidated. In this study, we utilized public microarray data analysis and tissue microarray technologies to display that CD44 level was enhanced in breast cancer and was significantly correlated with histological grade and the status of estrogen receptor, progesterone receptor and human epidermal growth factor receptor-2 (HER2) and EGFR. Furthermore, mRNA expression of CD44 in breast tumors was positively correlated with basal cytokeratin markers KRT5 and KRT17, but inversely associated with luminal marker FOXA1. Besides, Kaplan-Meier analysis showed that high CD44 mRNA level had adverse impact on the progression-free survival of patients with HER2-expressing or basal-like breast cancer. Functionally, inhibition of EGFR activity by erlotinib impaired the invasion and migration ability of breast cancer cell lines. Western blot assays demonstrated that erlotinib treatment decreased the expression of CD44, accompanied with the reduced protein levels of mesenchymal and cancer stem cell markers. Collectively, this study suggested that the expression of CD44 was upregulated by EGFR pathway and CD44 had a robust impact on the development of breast cancer.
Collapse
Affiliation(s)
- Hanxiao Xu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Kongju Wu
- Nursing School of Pingdingshan University, Pingdingshan, Henan 467000, P.R. China
| | - Yijun Tian
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Qian Liu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Na Han
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xun Yuan
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Lu Zhang
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Gen Sheng Wu
- Departments of Oncology and Pathology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
43
|
Klobučar M, Sedić M, Gehrig P, Grossmann J, Bilić M, Kovač-Bilić L, Pavelić K, Kraljević Pavelić S. Basement membrane protein ladinin-1 and the MIF-CD44-β1 integrin signaling axis are implicated in laryngeal cancer metastasis. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1938-54. [PMID: 27460703 DOI: 10.1016/j.bbadis.2016.07.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 07/06/2016] [Accepted: 07/22/2016] [Indexed: 12/14/2022]
Abstract
Laryngeal squamous cell carcinoma (LSCC) is the most common form of malignant disease in the head and neck region characterized by frequent occurrence of metastases in the neck lymph nodes early in the disease onset. In the presented study, we performed quantitative proteomic profiling of patient-matched primary tumor and adjacent non-tumorous tissues derived from metastatic LSCC as to identify new protein candidates with potential diagnostic and therapeutic significance. Obtained results revealed for the first time involvement of the basement membrane protein ladinin-1 in laryngeal cancer metastases. Alterations in the cellular microenvironment that propel metastatic events in laryngeal cancer include activation of MIF-CD44-β1 integrin signal transduction pathway and induction of downstream signaling mediated by NF-κB and Src tyrosine kinase, which ultimately impinge on cytoskeletal dynamics and architecture resulting in increased cellular motility and invasiveness. In this context, particularly interesting finding is upregulation of several actin-binding proteins novel to laryngeal cancer pathogenesis including coronin-1C and plastin-2, whose functional significance in laryngeal carcinogenesis has yet to be established. We also detected for the first time a complete loss of afamin in metastatic laryngeal cancer tissues, which warrants further studies into its use as a possible marker for monitoring disease progression and/or treatment outcome.
Collapse
Affiliation(s)
- Marko Klobučar
- University of Rijeka, Department of Biotechnology, Centre for High-throughput Technologies, Radmile Matejčić 2, 51000 Rijeka, Croatia
| | - Mirela Sedić
- University of Rijeka, Department of Biotechnology, Centre for High-throughput Technologies, Radmile Matejčić 2, 51000 Rijeka, Croatia
| | - Peter Gehrig
- Functional Genomics Center Zürich, University of Zurich/ETH Zurich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Jonas Grossmann
- Functional Genomics Center Zürich, University of Zurich/ETH Zurich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Mario Bilić
- University Department of ENT, Head and Neck Surgery, Kišpatićeva 12, 10000 Zagreb, Croatia
| | - Lana Kovač-Bilić
- University Department of ENT, Head and Neck Surgery, Kišpatićeva 12, 10000 Zagreb, Croatia
| | - Krešimir Pavelić
- University of Rijeka, Department of Biotechnology, Centre for High-throughput Technologies, Radmile Matejčić 2, 51000 Rijeka, Croatia
| | - Sandra Kraljević Pavelić
- University of Rijeka, Department of Biotechnology, Centre for High-throughput Technologies, Radmile Matejčić 2, 51000 Rijeka, Croatia.
| |
Collapse
|
44
|
Yin L, Fang F, Song X, Wang Y, Huang G, Su J, Hui N, Lu J. The pro-adhesive and pro-survival effects of glucocorticoid in human ovarian cancer cells. J Mol Endocrinol 2016; 57:61-72. [PMID: 27151574 DOI: 10.1530/jme-15-0142] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Accepted: 05/04/2016] [Indexed: 12/24/2022]
Abstract
Cell adhesion to extracellular matrix (ECM) is controlled by multiple signaling molecules and intracellular pathways, and is pivotal for survival and growth of cells from most solid tumors. Our previous works demonstrated that dexamethasone (DEX) significantly enhances cell adhesion and cell resistance to chemotherapeutics by increasing the levels of integrin β1, α4, and α5 in human ovarian cancer cells. However, it is unclear whether the components of ECM or other membrane molecules are also involved in the pro-adhesive effect of DEX in ovarian cancer cells. In this study, we demonstrated that the treatment of cells with DEX did not change the expression of collagens (I, III, and IV), laminin, CD44, and its principal ligand hyaluronan (HA), but significantly increased the levels of intracellular and secreted fibronectin (FN). Inhibiting the expression of FN with FN1 siRNA or blocking CD44, another FN receptor, with CD44 blocking antibody significantly attenuated the pro-adhesion of DEX, indicating that upregulation of FN mediates the pro-adhesive effect of DEX by its interaction with CD44 besides integrin β1. Moreover, DEX significantly enhanced cell resistance to the chemotherapeutic agent paclitaxel (PTX) by activating PI-3K-Akt pathway. Finally, we found that DEX also significantly upregulated the expression of MUC1, a transmembrane glycoprotein. Inhibiting the expression of MUC1 with MUC1 siRNA significantly attenuated the DEX-induced effects of pro-adhesion, Akt-activation, and pro-survival. In conclusion, these results provide new data that upregulation of FN and MUC1 by DEX contributes to DEX-induced pro-adhesion and protects ovarian cancer cells from chemotherapy.
Collapse
Affiliation(s)
- Lijuan Yin
- Department of PathophysiologyThe Second Military Medical University, Shanghai, China
| | - Fang Fang
- Department of Obstetrics and GynecologyChanghai Hospital, The Second Military Medical University, Shanghai, China
| | - Xinglei Song
- Department of PathophysiologyThe Second Military Medical University, Shanghai, China
| | - Yan Wang
- Department of PathophysiologyThe Second Military Medical University, Shanghai, China
| | - Gaoxiang Huang
- Department of PathophysiologyThe Second Military Medical University, Shanghai, China
| | - Jie Su
- Department of PathophysiologyThe Second Military Medical University, Shanghai, China
| | - Ning Hui
- Department of Obstetrics and GynecologyChanghai Hospital, The Second Military Medical University, Shanghai, China
| | - Jian Lu
- Department of PathophysiologyThe Second Military Medical University, Shanghai, China
| |
Collapse
|
45
|
Xu H, Tian Y, Yuan X, Wu H, Liu Q, Pestell RG, Wu K. The role of CD44 in epithelial-mesenchymal transition and cancer development. Onco Targets Ther 2015; 8:3783-92. [PMID: 26719706 PMCID: PMC4689260 DOI: 10.2147/ott.s95470] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
CD44, a multi-structural and multifunctional transmembrane glycoprotein, was initially identified as a receptor for hyaluronan that participates in both physiological and pathological processes. CD44 is found to be closely linked to the development of various solid tumors. Molecular studies have revealed that high CD44 expression was correlated with the phenotypes of cancer stem cells and epithelial–mesenchymal transition, thereby contributing to tumor invasion, metastasis, recurrence, and chemoresistance. Correspondingly, blockade of CD44 has been demonstrated to be capable of attenuating the malignant phenotype, slowing cancer progression, and reversing therapy resistance. Clinical analyses showed that high CD44 expression is associated with poor survival of various cancer patients, indicating that CD44 can be a potential prognostic marker. In this review, we summarize recent research progress of CD44 on tumor biology and the clinical significance of CD44.
Collapse
Affiliation(s)
- Hanxiao Xu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yijun Tian
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xun Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Hua Wu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Qian Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Richard G Pestell
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kongming Wu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|