1
|
Wang F, Yu J, Lu X, Numata K, Ruan L, Zhang D, Liu X, Li X, Wan M, Zhang W, Zhang G. Relationship between contrast-enhanced ultrasound combined with ultrasound resolution microscopy imaging and histological features of hepatocellular carcinoma. Abdom Radiol (NY) 2025:10.1007/s00261-025-04825-y. [PMID: 39928101 DOI: 10.1007/s00261-025-04825-y] [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: 11/24/2024] [Revised: 01/20/2025] [Accepted: 01/25/2025] [Indexed: 02/11/2025]
Abstract
OBJECTIVES Using contrast-enhanced ultrasound (CEUS) and ultrasound resolution microscopy (URM) imaging, this study aimed to evaluate the relationship between microvascular parameters of small hepatocellular carcinoma (sHCC) (≤ 3 cm) and microscopic histological features, which include vessels encapsulating tumour clusters (VETC), microvascular invasion (MVI), and histological grade. METHODS Sixteen patients with solitary resected sHCC were prospectively enrolled. CEUS and URM were performed one week before resection. All "ratio" refers to comparisons between the active area (where CEUS microbubble show visible motion track by URM) and the entire lesion. Blood vessel complexity (ratio), blood vessel density (ratio), area (ratio), flow velocity, blood vessel diameter, and perfusion index ("flow velocity" × "vessel ratio") were analysed using URM. The relationships between URM parameters and microscopic histological features (MVI, VETC, and histological grade) were analysed. RESULTS There were 5 (31.3%), 8 (50%), and 7 (43.7%) cases of poorly differentiated, MVI-positive, and VETC-positive HCC, respectively. The mean velocity of the entire lesion was higher in the poorly differentiated group than that in the moderately differentiated group (p = 0.026). The complexity ratio (MVI-positive: 1.07 ± 0.03, MVI-negative: 1.03 ± 0.02, p = 0.012), area ratio (MVI-positive: 0.63 ± 0.18, MVI-negative: 0.39 ± 0.16, p = 0.017), and perfusion index (MVI-positive: 8.67 ± 1.88, MVI-negative: 6.42 ± 0.94, p = 0.009) were greater in MVI-positive HCCs. The density ratio (VETC-positive: 1.30 ± 0.19, VETC-negative: 1.10 ± 0.05, p = 0.006) was larger in VETC-positive HCCs. CONCLUSION Higher blood flow velocity and area of HCC lesions, and higher blood vessel complexity and density may be related to microscopic histological features. This relationship might provide a strategy of using URM for preoperative non-invasive diagnostic VETC, MVI, and histological grade in the future.
Collapse
Affiliation(s)
- Feiqian Wang
- First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Jingtong Yu
- First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Baoji Hospital of Traditional Chinese Medicine, Baoji, China
| | - Xingqi Lu
- First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Baoji Hospital of Traditional Chinese Medicine, Baoji, China
| | - Kazushi Numata
- Yokohama City University Medical Center, Yokohama, Japan
| | - Litao Ruan
- First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Dong Zhang
- First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xi Liu
- First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaojing Li
- First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | | | - Wenbin Zhang
- VINNO Technology Company Limited, Jiangsu, China
| | - Guanjun Zhang
- First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
2
|
Saadh MJ, Bishoyi AK, Ballal S, Singh A, Kareem RA, Devi A, Sharma GC, Naidu KS, Sead FF. MicroRNAs as behind-the-scenes molecules in breast cancer metastasis and their therapeutic role through novel microRNA-based delivery strategies. Gene 2025; 944:149272. [PMID: 39894085 DOI: 10.1016/j.gene.2025.149272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
Breast cancer is the primary cause of cancer-related death and the most frequent malignancy among women in Western countries. Although there have been advancements in combination treatments and targeted therapies for the metastatic diseases management, metastatic breast cancer is still the second most common cause of cancer-related deaths among U.S. women. The routes of metastasis encompass invasion, intravasation, circulation, extravasation, infiltration into a remote location to establish a metastatic niche, and the formation of micro-metastases in a new environment. Each of these processes is regulated by changes in gene expression. MicroRNAs (miRNAs) are widely expressed by a variety of organisms and have a key role in cell activities including suppressing or promoting cancer through regulating various pathways. Target gene expression is post-transcriptionally regulated by miRNAs, which contribute to the development, spread, and metastasis of breast cancer. In this study, we comprehensively discussed the role of miRNAs as predictors of breast cancer metastasis, their correlation with the spread of the disease to certain organs, and their potential application as targets for breast cancer treatment. We also provided molecular mechanisms of miRNAs in the progression of breast cancer, as well as current challenges in miRNA-based therapeutic approaches. Furthermore, as one of the primary issues with the treatment of solid malignancies is the efficient delivery of miRNAs, we examined a number of cutting-edge carriers for miRNA-based therapies and CRISPR/Cas9 as a targeted therapy for breast cancer.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan.
| | - Ashok Kumar Bishoyi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot 360003, Gujarat, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401, Punjab, India
| | | | - Anita Devi
- Department of Chemistry Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | - Girish Chandra Sharma
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Fadhil Faez Sead
- Department of Dentistry, College of Dentistry, The Islamic University, Najaf, Iraq; Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
| |
Collapse
|
3
|
Kabak EC, Foo SL, Rafaeva M, Martin I, Bentires-Alj M. Microenvironmental Regulation of Dormancy in Breast Cancer Metastasis: "An Ally that Changes Allegiances". ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:373-395. [PMID: 39821034 DOI: 10.1007/978-3-031-70875-6_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Breast cancer remission after treatment is sometimes long-lasting, but in about 30% of cases, there is a relapse after a so-called dormant state. Cellular cancer dormancy, the propensity of disseminated tumor cells (DTCs) to remain in a nonproliferative state for an extended period, presents an opportunity for therapeutic intervention that may prevent reawakening and the lethal consequences of metastatic outgrowth. Therefore, identification of dormant DTCs and detailed characterization of cancer cell-intrinsic and niche-specific [i.e., tumor microenvironment (TME) mediated] mechanisms influencing dormancy in different metastatic organs are of great importance in breast cancer. Several microenvironmental drivers of DTC dormancy in metastatic organs, such as the lung, bone, liver, and brain, have been identified using in vivo models and/or in vitro three-dimensional culture systems. TME induction and persistence of dormancy in these organs are mainly mediated by signals from immune cells, stromal cells, and extracellular matrix components of the TME. Alterations of the TME have been shown to reawaken dormant DTCs. Efforts to capitalize on these findings often face translational challenges due to limited availability of representative patient samples and difficulty in designing dormancy-targeting clinical trials. In this chapter, we discuss current approaches to identify dormant DTCs and provide insights into cell-extrinsic (i.e., TME) mechanisms driving breast cancer cell dormancy in distant organs.
Collapse
Affiliation(s)
- Evrim Ceren Kabak
- Laboratory of Tumor Heterogeneity, Metastasis and Resistance, Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Sok Lin Foo
- Laboratory of Tumor Heterogeneity, Metastasis and Resistance, Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - Maria Rafaeva
- Laboratory of Tumor Heterogeneity, Metastasis and Resistance, Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Mohamed Bentires-Alj
- Laboratory of Tumor Heterogeneity, Metastasis and Resistance, Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
4
|
Chang CJ, Kan KW, Shih YY, Nan YH, Juan YC, Tu CH, Yang CT, Hsu WL, Cho DY, Shie MY, Chen YW, Chen CC. Preliminary study of utilizing a patient derived tumor spheroid model to augment precision therapy in metastatic brain tumors. Sci Rep 2024; 14:31888. [PMID: 39738713 DOI: 10.1038/s41598-024-83409-0] [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: 04/30/2024] [Accepted: 12/13/2024] [Indexed: 01/02/2025] Open
Abstract
Treating metastatic brain tumors remains a significant challenge. This study introduces and applies the Patient-Derived Tumor Spheroid (PDTS) system, an ex vivo model for precision drug testing on metastatic brain tumor. The PDTS system utilizes a decellularized extracellular matrix (dECM) derived from adipose tissue, combined with the tumor cells, to form tumor spheroids. These spheroids were subsequently used to test anticancer drugs, with results compared to the clinical outcomes observed after administering these treatments to patients. To assess the validity of the data, the correlation between the drug responses observed in the PDTS model and actual patient outcomes was analyzed. Chi-square tests evaluated the significance of associations between lab predictions and clinical outcomes, using a significance threshold of p < 0.05. In preliminary data, 17 patients met the criteria for final analysis, which showed an overall 57% accuracy (p-value = 0.463), with improvements to 73% accuracy (p-value = 0.072) when patients receiving certain treatments were excluded. This PDTS offers real-time results within three weeks, simultaneous testing of multiple drugs, and the ability to culture and store tumor cells for reproducibility. Despite some limitations, further development of this model could enhance its clinical application and improve patient outcomes.
Collapse
Affiliation(s)
- Chun-Jen Chang
- Department of Neurosurgery, China Medical University Hospital, 2 Hsueh‑Shih Road, Taichung City, 40402, Taiwan, ROC
| | - Kai-Wen Kan
- x-Dimension Center for Medical Research and Translation, China Medical University Hospital, Taichung, 404332, Taiwan
| | - Yu-Yin Shih
- Research and Development Center for x-Dimensional Extracellular Vesicles, China Medical University Hospital, Taichung, 404332, Taiwan
| | - Yu-Han Nan
- Department of Neurosurgery, China Medical University Hospital, 2 Hsueh‑Shih Road, Taichung City, 40402, Taiwan, ROC
| | - Yu-Chung Juan
- Department of Neurosurgery, China Medical University Hospital, 2 Hsueh‑Shih Road, Taichung City, 40402, Taiwan, ROC
| | - Chih-Hsiu Tu
- Department of Neurosurgery, China Medical University Hospital, 2 Hsueh‑Shih Road, Taichung City, 40402, Taiwan, ROC
| | - Chen-Ting Yang
- Department of Neurosurgery, China Medical University Hospital, 2 Hsueh‑Shih Road, Taichung City, 40402, Taiwan, ROC
| | - Wei-Lin Hsu
- Department of Neurosurgery, China Medical University Hospital, 2 Hsueh‑Shih Road, Taichung City, 40402, Taiwan, ROC
| | - Der-Yang Cho
- Department of Neurosurgery, China Medical University Hospital, 2 Hsueh‑Shih Road, Taichung City, 40402, Taiwan, ROC
| | - Ming-You Shie
- x-Dimension Center for Medical Research and Translation, China Medical University Hospital, Taichung, 404332, Taiwan
- School of Dentistry, China Medical University, Taichung, 406040, Taiwan
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, 41354, Taiwan
| | - Yi-Wen Chen
- x-Dimension Center for Medical Research and Translation, China Medical University Hospital, Taichung, 404332, Taiwan
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, 41354, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung City, 406040, Taiwan
| | - Chun-Chung Chen
- Department of Neurosurgery, China Medical University Hospital, 2 Hsueh‑Shih Road, Taichung City, 40402, Taiwan, ROC.
- Graduate Institute of Acupuncture Science, China Medical University, Taichung City, 40402, Taiwan, ROC.
- Neuroscience and Brain Disease Center, China Medical University, Li-fu Hall 8F, No. 91, Xue Shi Rd., North Dist., Taichung City, 40402, Taiwan, ROC.
- School of Medicine, China Medical University, Taichung, Taiwan.
- Department of Neurosurgery, China Medical University Hospital, 2, Yu-Der Road, Taichung, 40447, Taiwan.
| |
Collapse
|
5
|
Vrynas A, Bazban-Shotorbani S, Arfan S, Satia K, Ashna M, Zhang A, Visan D, Chen A, Carter M, Blackhall F, Simpson KL, Dive C, Huang P, Au SH. Circulating tumor cells shed large extracellular vesicles in capillary bifurcations that activate endothelial and immune cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.17.589880. [PMID: 38659882 PMCID: PMC11042361 DOI: 10.1101/2024.04.17.589880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Circulating tumor cells (CTCs) and their clusters are the drivers of metastasis, but we have an incomplete understanding of how they interact with capillary beds. Using microfluidic models mimicking human capillary bifurcations, we observed cell size- and bifurcation-dependent shedding of nuclei-free fragments by patient CTCs, CTC-derived explant cells and numerous cancer cell lines. Shedding reduced cell sizes up to 61%, facilitating their transit through bifurcations. We demonstrated that shed fragments were a novel subclass of large extracellular vesicles (LEVs), "shearosomes", that require shear stress for their biogenesis and whose proteome was associated with immune-related pathways. Shearosomes exhibited functions characteristic of previously identified EVs including cell-directed internalization by endothelial and immune cells, and intercellular communication abilities such as disruption of endothelial barrier integrity, polarization of monocytes into M2 tumor-promoting macrophages and interactions between endothelial and immune cells. Cumulatively, these findings suggest that CTCs shed shearosomes in capillary beds that drive key processes involved in the formation of pre-metastatic niches.
Collapse
Affiliation(s)
- Angelos Vrynas
- Department of Bioengineering, Imperial College London; London, SW7 2AZ, United Kingdom
| | | | - Sara Arfan
- Division of Molecular Pathology, The Institute of Cancer Research; London, SM2 5NG, United Kingdom
| | - Karishma Satia
- Cancer Research UK National Biomarker Centre, University of Manchester; Manchester, M13 9PL, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence; Manchester, M13 9PL, United Kingdom
| | - Mymuna Ashna
- Department of Bioengineering, Imperial College London; London, SW7 2AZ, United Kingdom
| | - Aoyu Zhang
- Department of Bioengineering, Imperial College London; London, SW7 2AZ, United Kingdom
| | - Diana Visan
- Department of Bioengineering, Imperial College London; London, SW7 2AZ, United Kingdom
| | - Aisher Chen
- Department of Bioengineering, Imperial College London; London, SW7 2AZ, United Kingdom
| | - Mathew Carter
- Cancer Research UK National Biomarker Centre, University of Manchester; Manchester, M13 9PL, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence; Manchester, M13 9PL, United Kingdom
- Medical Oncology, Christie Hospital National Health Service (NHS) Foundation Trust; Manchester, M20 4BX, United Kingdom
| | - Fiona Blackhall
- Cancer Research UK Lung Cancer Centre of Excellence; Manchester, M13 9PL, United Kingdom
- Medical Oncology, Christie Hospital National Health Service (NHS) Foundation Trust; Manchester, M20 4BX, United Kingdom
- The Division of Cancer Sciences, Faculty of Biology, Medicine, and Health, University of Manchester; Manchester, M13 9PL, United Kingdom
| | - Kathryn L Simpson
- Cancer Research UK National Biomarker Centre, University of Manchester; Manchester, M13 9PL, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence; Manchester, M13 9PL, United Kingdom
- SCLC Biology Group, Cancer Research UK Manchester Institute, University of Manchester; Manchester, M20 4BX, United Kingdom
| | - Caroline Dive
- Cancer Research UK National Biomarker Centre, University of Manchester; Manchester, M13 9PL, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence; Manchester, M13 9PL, United Kingdom
- SCLC Biology Group, Cancer Research UK Manchester Institute, University of Manchester; Manchester, M20 4BX, United Kingdom
| | - Paul Huang
- Division of Molecular Pathology, The Institute of Cancer Research; London, SM2 5NG, United Kingdom
- Cancer Research UK Convergence Science Centre; London, SW7 2AZ, United Kingdom
| | - Sam H Au
- Department of Bioengineering, Imperial College London; London, SW7 2AZ, United Kingdom
- Cancer Research UK Convergence Science Centre; London, SW7 2AZ, United Kingdom
| |
Collapse
|
6
|
Dupas A, Goetz JG, Osmani N. Extravasation of immune and tumor cells from an endothelial perspective. J Cell Sci 2024; 137:jcs262066. [PMID: 39530179 DOI: 10.1242/jcs.262066] [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: 11/16/2024] Open
Abstract
Crossing the vascular endothelium is a necessary stage for circulating cells aiming to reach distant organs. Leukocyte passage through the endothelium, known as transmigration, is a multistep process during which immune cells adhere to the vascular wall, migrate and crawl along the endothelium until they reach their exit site. Similarly, circulating tumor cells (CTCs), which originate from the primary tumor or reseed from early metastatic sites, disseminate using the blood circulation and also must cross the endothelial barrier to set new colonies in distant organs. CTCs are thought to mimic arrest and extravasation utilized by leukocytes; however, their extravasation also requires processes that, from an endothelial perspective, are specific to cancer cells. Although leukocyte extravasation relies on maintaining endothelial impermeability, it appears that cancer cells can indoctrinate endothelial cells into promoting their extravasation independently of their normal functions. In this Review, we summarize the common and divergent mechanisms of endothelial responses during extravasation of leukocytes (in inflammation) and CTCs (in metastasis), and highlight how these might be leveraged in the development of anti-metastatic treatments.
Collapse
Affiliation(s)
- Amandine Dupas
- Tumor Biomechanics lab, INSERM UMR_S 1109, CRBS, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg Cedex, France
- Université de Strasbourg, Strasbourg, F-67000, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, F-67000, France
- Equipe Labellisée Ligue Contre le Cancer, France
| | - Jacky G Goetz
- Tumor Biomechanics lab, INSERM UMR_S 1109, CRBS, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg Cedex, France
- Université de Strasbourg, Strasbourg, F-67000, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, F-67000, France
- Equipe Labellisée Ligue Contre le Cancer, France
| | - Naël Osmani
- Tumor Biomechanics lab, INSERM UMR_S 1109, CRBS, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg Cedex, France
- Université de Strasbourg, Strasbourg, F-67000, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, F-67000, France
- Equipe Labellisée Ligue Contre le Cancer, France
| |
Collapse
|
7
|
Sajib MS, Zahra FT, Lamprou M, Akwii RG, Park JH, Osorio M, Tullar P, Doci CL, Zhang C, Huveneers S, Van Buul JD, Wang MH, Markiewski MM, Srivastava SK, Zheng Y, Gutkind JS, Hu J, Bickel U, Maeda DY, Zebala JA, Lionakis MS, Trasti S, Mikelis CM. Tumor-induced endothelial RhoA activation mediates tumor cell transendothelial migration and metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.22.614304. [PMID: 39372784 PMCID: PMC11451620 DOI: 10.1101/2024.09.22.614304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
The endothelial barrier plays an active role in transendothelial tumor cell migration during metastasis, however, the endothelial regulatory elements of this step remain obscure. Here we show that endothelial RhoA activation is a determining factor during this process. Breast tumor cell-induced endothelial RhoA activation is the combined outcome of paracrine IL-8-dependent and cell-to-cell contact β 1 integrin-mediated mechanisms, with elements of this pathway correlating with clinical data. Endothelial-specific RhoA blockade or in vivo deficiency inhibited the transendothelial migration and metastatic potential of human breast tumor and three murine syngeneic tumor cell lines, similar to the pharmacological blockade of the downstream RhoA pathway. These findings highlight endothelial RhoA as a potent, universal target in the tumor microenvironment for anti-metastatic treatment of solid tumors.
Collapse
|
8
|
Li S, Feng T, Yuan H, Li Q, Zhao G, Li K. DEAD-box RNA helicases in the multistep process of tumor metastasis. Mol Biol Rep 2024; 51:1006. [PMID: 39306810 DOI: 10.1007/s11033-024-09912-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 09/03/2024] [Indexed: 01/04/2025]
Abstract
RNA helicases constitute a large family of proteins that share a catalytic core with high structural similarity. DEAD-box (DDX) proteins belong to the largest RNA helicase subfamily, and DDX members have been implicated in all facets of RNA metabolism, from transcription to translation, miRNA maturation, and RNA delay and degradation. Interestingly, an increasing number of studies have suggested a relationship between DDX proteins and cancer initiation and progression. The expression levels of many DDX proteins are elevated in a majority of cancers, and recent studies have demonstrated that some DDX proteins have a potent positive effect on promoting the metastasis of malignant cells. Metastasis is a complex, multistep cascade process that includes local invasion, intravasation and survival in the circulation, arrest at a distant organ site, extravasation and metastatic colonization; here, we review this process and present the suggested functions and mechanisms of DDX family proteins in particular steps of the invasion‒metastasis cascade.
Collapse
Affiliation(s)
- Shan Li
- Cancer Center and Lab of Experimental Oncology, State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu City, Sichuan Province, People's Republic of China
| | - Tianyu Feng
- Department of Laboratory Medicine, Clinical Laboratory Medicine Research Center of West China Hospital, Sichuan Clinical Research Center for Laboratory Medicine, West China Hospital, Sichuan University, Chengdu City, People's Republic of China
| | - Hang Yuan
- Cancer Center and Lab of Experimental Oncology, State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu City, Sichuan Province, People's Republic of China
| | - Qin Li
- Cancer Center and Lab of Experimental Oncology, State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu City, Sichuan Province, People's Republic of China
| | - Gang Zhao
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu City, Sichuan Province, People's Republic of China.
| | - Kai Li
- Cancer Center and Lab of Experimental Oncology, State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu City, Sichuan Province, People's Republic of China.
| |
Collapse
|
9
|
Shu Y, Li B, Ma H, Liu J, Cheng YY, Li X, Liu T, Yang C, Ma X, Song K. Three-dimensional breast cancer tumor models based on natural hydrogels: a review. J Zhejiang Univ Sci B 2024; 25:736-755. [PMID: 39308065 PMCID: PMC11422793 DOI: 10.1631/jzus.b2300840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Breast cancer is the most common cancer in women and one of the deadliest cancers worldwide. According to the distribution of tumor tissue, breast cancer can be divided into invasive and non-invasive forms. The cancer cells in invasive breast cancer pass through the breast and through the immune system or systemic circulation to different parts of the body, forming metastatic breast cancer. Drug resistance and distant metastasis are the main causes of death from breast cancer. Research on breast cancer has attracted extensive attention from researchers. In vitro construction of tumor models by tissue engineering methods is a common tool for studying cancer mechanisms and anticancer drug screening. The tumor microenvironment consists of cancer cells and various types of stromal cells, including fibroblasts, endothelial cells, mesenchymal cells, and immune cells embedded in the extracellular matrix. The extracellular matrix contains fibrin proteins (such as types I, II, III, IV, VI, and X collagen and elastin) and glycoproteins (such as proteoglycan, laminin, and fibronectin), which are involved in cell signaling and binding of growth factors. The current traditional two-dimensional (2D) tumor models are limited by the growth environment and often cannot accurately reproduce the heterogeneity and complexity of tumor tissues in vivo. Therefore, in recent years, research on three-dimensional (3D) tumor models has gradually increased, especially 3D bioprinting models with high precision and repeatability. Compared with a 2D model, the 3D environment can better simulate the complex extracellular matrix components and structures in the tumor microenvironment. Three-dimensional models are often used as a bridge between 2D cellular level experiments and animal experiments. Acellular matrix, gelatin, sodium alginate, and other natural materials are widely used in the construction of tumor models because of their excellent biocompatibility and non-immune rejection. Here, we review various natural scaffold materials and construction methods involved in 3D tissue-engineered tumor models, as a reference for research in the field of breast cancer.
Collapse
Affiliation(s)
- Yan Shu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Bing Li
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Hailin Ma
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Jiaqi Liu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Yuen Yee Cheng
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, NSW 2007, Australia
| | - Xiangqin Li
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Tianqing Liu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Chuwei Yang
- Emergency Center, the Second Hospital of Dalian Medical University, Dalian 116023, China. ,
| | - Xiao Ma
- Department of Anesthesia, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China. ,
| | - Kedong Song
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China.
| |
Collapse
|
10
|
Gasser E, Su E, Vaidžiulytė K, Abbade N, Cognart H, Manneville JB, Viovy JL, Piel M, Pierga JY, Terao K, Villard C. Deformation under flow and morphological recovery of cancer cells. LAB ON A CHIP 2024; 24:3930-3944. [PMID: 38993177 PMCID: PMC11302772 DOI: 10.1039/d4lc00246f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 06/30/2024] [Indexed: 07/13/2024]
Abstract
The metastatic cascade includes a blood circulation step for cells detached from the primary tumor. This stage involves significant shear stress as well as large and fast deformation as the cells circulate through the microvasculature. These mechanical stimuli are well reproduced in microfluidic devices. However, the recovery dynamics after deformation is also pivotal to understand how a cell can pass through the multiple capillary constrictions encountered during a single hemodynamic cycle. The microfluidic system developed in this work allows single cell recovery to be studied under flow-free conditions following pressure-actuated cell deformation inside constricted microchannels. We used three breast cancer cell lines - namely MCF-7, SK-BR3 and MDA-MB231 - as cellular models representative of different cancer phenotypes. Changing the size of the constriction allows exploration of moderate to strong deformation regimes, the latter being associated with the formation of plasma membrane blebs. In the regime of moderate deformation, all cell types display a fast elastic recovery behavior followed by a slower viscoelastic regime, well described by a double exponential decay. Among the three cell types, cells of the mesenchymal phenotype, i.e. the MDA-MB231 cells, are softer and the most fluid-like, in agreement with previous studies. Our main finding here is that the fast elastic recovery regime revealed by our novel microfluidic system is under the control of cell contractility ensured by the integrity of the cell cortex. Our results suggest that the cell cortex plays a major role in the transit of circulating tumor cells by allowing their fast morphological recovery after deformation in blood capillaries.
Collapse
Affiliation(s)
- Emile Gasser
- Institut Curie and Institut Pierre Gilles de Gennes, Physique des Cellules et Cancer, CNRS UMR168, Université PSL, F-75005 Paris, France.
- Laboratoire Interdisciplinaire des Energies de Demain, CNRS UMR 8236, Université Paris Cité, F-75013, Paris, France.
| | - Emilie Su
- Laboratoire Interdisciplinaire des Energies de Demain, CNRS UMR 8236, Université Paris Cité, F-75013, Paris, France.
- Laboratoire Matière et Systèmes Complexes (MSC), CNRS UMR 7057, Université Paris Cité, 10 Rue Alice Domon et Léonie Duquet, F-75013 Paris, France
| | - Kotryna Vaidžiulytė
- Institut Curie and Institut Pierre Gilles de Gennes, CNRS UMR144, Université PSL, F-75005 Paris, France
| | - Nassiba Abbade
- Institut Curie and Institut Pierre Gilles de Gennes, Physique des Cellules et Cancer, CNRS UMR168, Université PSL, F-75005 Paris, France.
- Institut Curie and Institut Pierre Gilles de Gennes, CNRS UMR144, Université PSL, F-75005 Paris, France
| | - Hamizah Cognart
- Institut Curie and Institut Pierre Gilles de Gennes, Physique des Cellules et Cancer, CNRS UMR168, Université PSL, F-75005 Paris, France.
| | - Jean-Baptiste Manneville
- Laboratoire Matière et Systèmes Complexes (MSC), CNRS UMR 7057, Université Paris Cité, 10 Rue Alice Domon et Léonie Duquet, F-75013 Paris, France
| | - Jean-Louis Viovy
- Institut Curie and Institut Pierre Gilles de Gennes, Physique des Cellules et Cancer, CNRS UMR168, Université PSL, F-75005 Paris, France.
| | - Matthieu Piel
- Institut Curie and Institut Pierre Gilles de Gennes, CNRS UMR144, Université PSL, F-75005 Paris, France
| | - Jean-Yves Pierga
- Département d'Oncologie Médicale de l'Institut Curie et Université Paris Cité, France
| | - Kyohei Terao
- Nano-Micro Structure Device Integrated Research Center, Kagawa University, 2217-20 Hayashi-cho, Takamatsu 761-0396, Japan.
| | - Catherine Villard
- Laboratoire Interdisciplinaire des Energies de Demain, CNRS UMR 8236, Université Paris Cité, F-75013, Paris, France.
| |
Collapse
|
11
|
He J, Duan P, Liu Y, Feng T, Wang S, Lin X, Xie J, Liu X. Unveiling the Impact of Hemodynamics on Endothelial Inflammation-Mediated Hepatocellular Carcinoma Metastasis Using a Biomimetic Vascular Flow Model. Adv Healthc Mater 2024; 13:e2304439. [PMID: 38486060 DOI: 10.1002/adhm.202304439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/28/2024] [Indexed: 03/23/2024]
Abstract
Hepatocellular carcinoma (HCC) hematogenous dissemination is a leading cause of HCC-related deaths. The inflammatory facilitates this process by promoting the adhesion and invasion of tumor cells in the circulatory system. But the contribution of hemodynamics to this process remains poorly understood due to the lack of a suitable vascular flow model for investigation. This study develops a vascular flow model to examine the impact of hemodynamics on endothelial inflammation-mediated HCC metastasis. This work finds the increasing shear stress will reduce the recruitment of HCC cells by disturbing adhesion forces between endothelium and HCC cells. However, this reduction will be restored by the inflammation. When applying high FSS (4-6 dyn cm-2) to the inflammatory endothelium, there will be a 4.8-fold increase in HCC cell adhesions compared to normal condition. Nevertheless, the increase fold of cell adhesions is inapparent, around 1.5-fold, with low and medium FSS. This effect can be attributed to the FSS-induced upregulation of ICAM-1 and VCAM-1 of the inflammatory endothelium, which serve to strengthen cell binding forces. These findings indicate that hemodynamics plays a key role in HCC metastasis during endothelial inflammation by regulating the expression of adhesion-related factors.
Collapse
Affiliation(s)
- Jia He
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Peiyan Duan
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Yi Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Tang Feng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shuo Wang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Xinyi Lin
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Jing Xie
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
12
|
Mahasa KJ, Ouifki R, de Pillis L, Eladdadi A. A Role of Effector CD 8 + T Cells Against Circulating Tumor Cells Cloaked with Platelets: Insights from a Mathematical Model. Bull Math Biol 2024; 86:89. [PMID: 38884815 DOI: 10.1007/s11538-024-01323-y] [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: 01/18/2024] [Accepted: 05/31/2024] [Indexed: 06/18/2024]
Abstract
Cancer metastasis accounts for a majority of cancer-related deaths worldwide. Metastasis occurs when the primary tumor sheds cells into the blood and lymphatic circulation, thereby becoming circulating tumor cells (CTCs) that transverse through the circulatory system, extravasate the circulation and establish a secondary distant tumor. Accumulating evidence suggests that circulating effector CD 8 + T cells are able to recognize and attack arrested or extravasating CTCs, but this important antitumoral effect remains largely undefined. Recent studies highlighted the supporting role of activated platelets in CTCs's extravasation from the bloodstream, contributing to metastatic progression. In this work, a simple mathematical model describes how the primary tumor, CTCs, activated platelets and effector CD 8 + T cells participate in metastasis. The stability analysis reveals that for early dissemination of CTCs, effector CD 8 + T cells can present or keep secondary metastatic tumor burden at low equilibrium state. In contrast, for late dissemination of CTCs, effector CD 8 + T cells are unlikely to inhibit secondary tumor growth. Moreover, global sensitivity analysis demonstrates that the rate of the primary tumor growth, intravascular CTC proliferation, as well as the CD 8 + T cell proliferation, strongly affects the number of the secondary tumor cells. Additionally, model simulations indicate that an increase in CTC proliferation greatly contributes to tumor metastasis. Our simulations further illustrate that the higher the number of activated platelets on CTCs, the higher the probability of secondary tumor establishment. Intriguingly, from a mathematical immunology perspective, our simulations indicate that if the rate of effector CD 8 + T cell proliferation is high, then the secondary tumor formation can be considerably delayed, providing a window for adjuvant tumor control strategies. Collectively, our results suggest that the earlier the effector CD 8 + T cell response is enhanced the higher is the probability of preventing or delaying secondary tumor metastases.
Collapse
Affiliation(s)
- Khaphetsi Joseph Mahasa
- Department of Mathematics and Computer Science, National University of Lesotho, Roma, Maseru, Lesotho.
| | - Rachid Ouifki
- Department of Mathematics and Applied Mathematics, Mafikeng Campus, North-West University, Private Bag X2046, Mmabatho, 2735, South Africa
| | | | - Amina Eladdadi
- Division of Mathematical Sciences, The National Science Foundation, Alexandria, VA, USA
| |
Collapse
|
13
|
Xin Y, Hu B, Li K, Hu G, Zhang C, Chen X, Tang K, Du P, Tan Y. Circulating tumor cells with metastasis-initiating competence survive fluid shear stress during hematogenous dissemination through CXCR4-PI3K/AKT signaling. Cancer Lett 2024; 590:216870. [PMID: 38614386 DOI: 10.1016/j.canlet.2024.216870] [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: 01/11/2024] [Revised: 03/28/2024] [Accepted: 04/06/2024] [Indexed: 04/15/2024]
Abstract
To seed lethal secondary lesions, circulating tumor cells (CTCs) must survive all rate-limiting factors during hematogenous dissemination, including fluid shear stress (FSS) that poses a grand challenge to their survival. We thus hypothesized that CTCs with the ability to survive FSS in vasculature might hold metastasis-initiating competence. This study reported that FSS of physiologic magnitude selected a small subpopulation of suspended tumor cells in vitro with the traits of metastasis-initiating cells, including stemness, migration/invasion potential, cellular plasticity, and biophysical properties. These shear-selected cells generated local and metastatic tumors at the primary and distal sites efficiently, implicating their metastasis competence. Mechanistically, FSS activated the mechanosensitive protein CXCR4 and the downstream PI3K/AKT signaling, which were essential in shear-mediated selection of metastasis-competent CTCs. In summary, these findings conclude that CTCs with metastasis-initiating competence survive FSS during hematogenous dissemination through CXCR4-PI3K/AKT signaling, which may provide new therapeutic targets for the early prevention of tumor metastasis.
Collapse
Affiliation(s)
- Ying Xin
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China; Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, 999077, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Bing Hu
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China; Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, 999077, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Keming Li
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China; Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Guanshuo Hu
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China; Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, 999077, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Cunyu Zhang
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China; Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, 999077, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Xi Chen
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China; Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Kai Tang
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China; Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Pengyu Du
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China; Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Youhua Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China; Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, 999077, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, 999077, China.
| |
Collapse
|
14
|
Jouybar M, de Winde CM, Wolf K, Friedl P, Mebius RE, den Toonder JMJ. Cancer-on-chip models for metastasis: importance of the tumor microenvironment. Trends Biotechnol 2024; 42:431-448. [PMID: 37914546 DOI: 10.1016/j.tibtech.2023.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 11/03/2023]
Abstract
Cancer-on-chip (CoC) models, based on microfluidic chips harboring chambers for 3D tumor-cell culture, enable us to create a controlled tumor microenvironment (TME). CoC models are therefore increasingly used to systematically study effects of the TME on the various steps in cancer metastasis. Moreover, CoC models have great potential for developing novel cancer therapies and for predicting patient-specific response to cancer treatments. We review recent developments in CoC models, focusing on three main TME components: (i) the anisotropic extracellular matrix (ECM) architectures, (ii) the vasculature, and (iii) the immune system. We aim to provide guidance to biologists to choose the best CoC approach for addressing questions about the role of the TME in metastasis, and to inspire engineers to develop novel CoC technologies.
Collapse
Affiliation(s)
- Mohammad Jouybar
- Microsystems, Eindhoven University of Technology, Eindhoven, The Netherlands; Institute for Complex Molecular Systems, Eindhoven, The Netherlands
| | - Charlotte M de Winde
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology & Immunology, Amsterdam, The Netherlands; Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands; Cancer Center Amsterdam, Cancer Biology & Immunology, Amsterdam, The Netherlands
| | - Katarina Wolf
- Department of Medical BioSciences, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter Friedl
- Department of Medical BioSciences, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Cancer Genomics Center, Utrecht, The Netherlands
| | - Reina E Mebius
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology & Immunology, Amsterdam, The Netherlands; Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands; Cancer Center Amsterdam, Cancer Biology & Immunology, Amsterdam, The Netherlands; Amsterdam Institute for Infection and Immunity, Inflammatory diseases, Amsterdam, The Netherlands
| | - Jaap M J den Toonder
- Microsystems, Eindhoven University of Technology, Eindhoven, The Netherlands; Institute for Complex Molecular Systems, Eindhoven, The Netherlands.
| |
Collapse
|
15
|
Lee M, Kim S, Lee SY, Son JG, Park J, Park S, Yeun J, Lee TG, Im SG, Jeon JS. Hydrophobic surface induced pro-metastatic cancer cells for in vitro extravasation models. Bioact Mater 2024; 34:401-413. [PMID: 38282966 PMCID: PMC10819557 DOI: 10.1016/j.bioactmat.2023.12.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 12/08/2023] [Accepted: 12/23/2023] [Indexed: 01/30/2024] Open
Abstract
In vitro vascularized cancer models utilizing microfluidics have emerged as a promising tool for mechanism study and drug screening. However, the lack of consideration and preparation methods for cancer cellular sources that are capable of adequately replicating the metastatic features of circulating tumor cells contributed to low relevancy with in vivo experimental results. Here, we show that the properties of cancer cellular sources have a considerable impact on the validity of the in vitro metastasis model. Notably, with a hydrophobic surface, we can create highly metastatic spheroids equipped with aggressive invasion, endothelium adhesion capabilities, and activated metabolic features. Combining these metastatic spheroids with the well-constructed microfluidic-based extravasation model, we validate that these metastatic spheroids exhibited a distinct extravasation response to epidermal growth factor (EGF) and normal human lung fibroblasts compared to the 2D cultured cancer cells, which is consistent with the previously reported results of in vivo experiments. Furthermore, the applicability of the developed model as a therapeutic screening platform for cancer extravasation is validated through profiling and inhibition of cytokines. We believe this model incorporating hydrophobic surface-cultured 3D cancer cells provides reliable experimental data in a clear and concise manner, bridging the gap between the conventional in vitro models and in vivo experiments.
Collapse
Affiliation(s)
- Minseok Lee
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daehak-ro 291, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Seunggyu Kim
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daehak-ro 291, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Sun Young Lee
- Bioimaging Team, Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), Gajeong-ro 267, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Jin Gyeong Son
- Bioimaging Team, Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), Gajeong-ro 267, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Joonha Park
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daehak-ro 291, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Seonghyeon Park
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daehak-ro 291, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Jemin Yeun
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daehak-ro 291, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Tae Geol Lee
- Bioimaging Team, Safety Measurement Institute, Korea Research Institute of Standards and Science (KRISS), Gajeong-ro 267, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Sung Gap Im
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daehak-ro 291, Yuseong-gu, Daejeon, 34141, Republic of Korea
- KAIST Institute for the NanoCentury (KINC), Korea Advanced Institute of Science and Technology, Daehak-ro 291, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Jessie S. Jeon
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daehak-ro 291, Yuseong-gu, Daejeon, 34141, Republic of Korea
| |
Collapse
|
16
|
Keshavarz Motamed P, Abouali H, Poudineh M, Maftoon N. Experimental measurement and numerical modeling of deformation behavior of breast cancer cells passing through constricted microfluidic channels. MICROSYSTEMS & NANOENGINEERING 2024; 10:7. [PMID: 38222473 PMCID: PMC10786721 DOI: 10.1038/s41378-023-00644-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 11/11/2023] [Accepted: 11/27/2023] [Indexed: 01/16/2024]
Abstract
During the multistep process of metastasis, cancer cells encounter various mechanical forces which make them deform drastically. Developing accurate in-silico models, capable of simulating the interactions between the mechanical forces and highly deformable cancer cells, can pave the way for the development of novel diagnostic and predictive methods for metastatic progression. Spring-network models of cancer cell, empowered by our recently proposed identification approach, promises a versatile numerical tool for developing experimentally validated models that can simulate complex interactions at cellular scale. Using this numerical tool, we presented spring-network models of breast cancer cells that can accurately replicate the experimental data of deformation behavior of the cells flowing in a fluidic domain and passing narrow constrictions comparable to microcapillary. First, using high-speed imaging, we experimentally studied the deformability of breast cancer cell lines with varying metastatic potential (MCF-7 (less invasive), SKBR-3 (medium-high invasive), and MDA-MB-231 (highly invasive)) in terms of their entry time to a constricted microfluidic channel. We observed that MDA-MB-231, that has the highest metastatic potential, is the most deformable cell among the three. Then, by focusing on this cell line, experimental measurements were expanded to two more constricted microchannel dimensions. The experimental deformability data in three constricted microchannel sizes for various cell sizes, enabled accurate identification of the unknown parameters of the spring-network model of the breast cancer cell line (MDA-MB-231). Our results show that the identified parameters depend on the cell size, suggesting the need for a systematic procedure for identifying the size-dependent parameters of spring-network models of cells. As the numerical results show, the presented cell models can simulate the entry process of the cell into constricted channels with very good agreements with the measured experimental data.
Collapse
Affiliation(s)
- Pouyan Keshavarz Motamed
- Department of Systems Design Engineering, University of Waterloo, Waterloo, ON N2L 3G1 Canada
- Center for Bioengineering and Biotechnology, University of Waterloo, Waterloo, ON N2L 3G1 Canada
| | - Hesam Abouali
- Center for Bioengineering and Biotechnology, University of Waterloo, Waterloo, ON N2L 3G1 Canada
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON N2L 3G1 Canada
| | - Mahla Poudineh
- Center for Bioengineering and Biotechnology, University of Waterloo, Waterloo, ON N2L 3G1 Canada
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON N2L 3G1 Canada
| | - Nima Maftoon
- Department of Systems Design Engineering, University of Waterloo, Waterloo, ON N2L 3G1 Canada
- Center for Bioengineering and Biotechnology, University of Waterloo, Waterloo, ON N2L 3G1 Canada
| |
Collapse
|
17
|
Castillo SP, Rebolledo RA, Arim M, Hochberg ME, Marquet PA. Metastatic cells exploit their stoichiometric niche in the network of cancer ecosystems. SCIENCE ADVANCES 2023; 9:eadi7902. [PMID: 38091399 PMCID: PMC10848726 DOI: 10.1126/sciadv.adi7902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 11/10/2023] [Indexed: 12/18/2023]
Abstract
Metastasis is a nonrandom process with varying degrees of organotropism-specific source-acceptor seeding. Understanding how patterns between source and acceptor tumors emerge remains a challenge in oncology. We hypothesize that organotropism results from the macronutrient niche of cells in source and acceptor organs. To test this, we constructed and analyzed a metastatic network based on 9303 records across 28 tissue types. We found that the topology of the network is nested and modular with scale-free degree distributions, reflecting organotropism along a specificity/generality continuum. The variation in topology is significantly explained by the matching of metastatic cells to their stoichiometric niche. Specifically, successful metastases are associated with higher phosphorus content in the acceptor compared to the source organ, due to metabolic constraints in proliferation crucial to the invasion of new tissues. We conclude that metastases are codetermined by processes at source and acceptor organs, where phosphorus content is a limiting factor orchestrating tumor ecology.
Collapse
Affiliation(s)
- Simon P. Castillo
- Departamento de Ecología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, C.P. 8331150, Santiago, Chile
| | - Rolando A. Rebolledo
- Instituto de Ingeniería Biológica y Médica (IIBM), Pontificia Universidad Católica de Chile, Santiago, Chile
- Hepato-Pancreato-Biliary Surgery Unit, Surgery Service, Complejo Asistencial Dr. Sótero Del Río, Santiago, Chile
| | - Matías Arim
- Departamento de Ecologia y Gestion Ambiental, Centro Universitario Regional Este (CURE), Universidad de la República, Maldonado, Uruguay
| | - Michael E. Hochberg
- ISEM, University of Montpellier, Montpellier, France
- Santa Fe Institute, Santa Fe, NM 87501, USA
| | - Pablo A. Marquet
- Departamento de Ecología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, C.P. 8331150, Santiago, Chile
- Santa Fe Institute, Santa Fe, NM 87501, USA
- Centro de Modelamiento Matemático, Universidad de Chile, International Research Laboratory 2807, CNRS, C.P. 8370456, Santiago, Chile
- Instituto de Sistemas Complejos de Valparaíso (ISCV), Valparaíso, Chile
| |
Collapse
|
18
|
Gounou C, Rouyer L, Siegfried G, Harté E, Bouvet F, d'Agata L, Darbo E, Lefeuvre M, Derieppe MA, Bouton L, Mélane M, Chapeau D, Martineau J, Prouzet-Mauleon V, Tan S, Souleyreau W, Saltel F, Argoul F, Khatib AM, Brisson AR, Iggo R, Bouter A. Inhibition of the membrane repair protein annexin-A2 prevents tumor invasion and metastasis. Cell Mol Life Sci 2023; 81:7. [PMID: 38092984 PMCID: PMC10719157 DOI: 10.1007/s00018-023-05049-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 11/09/2023] [Accepted: 11/11/2023] [Indexed: 12/17/2023]
Abstract
Cancer cells are exposed to major compressive and shearing forces during invasion and metastasis, leading to extensive plasma membrane damage. To survive this mechanical stress, they need to repair membrane injury efficiently. Targeting the membrane repair machinery is thus potentially a new way to prevent invasion and metastasis. We show here that annexin-A2 (ANXA2) is required for membrane repair in invasive breast and pancreatic cancer cells. Mechanistically, we show by fluorescence and electron microscopy that cells fail to reseal shear-stress damaged membrane when ANXA2 is silenced or the protein is inhibited with neutralizing antibody. Silencing of ANXA2 has no effect on proliferation in vitro, and may even accelerate migration in wound healing assays, but reduces tumor cell dissemination in both mice and zebrafish. We expect that inhibiting membrane repair will be particularly effective in aggressive, poor prognosis tumors because they rely on the membrane repair machinery to survive membrane damage during tumor invasion and metastasis. This could be achieved either with anti-ANXA2 antibodies, which have been shown to inhibit metastasis of breast and pancreatic cancer cells, or with small molecule drugs.
Collapse
Affiliation(s)
- C Gounou
- CNRS, Bordeaux INP, CBMN, UMR 5248, University of Bordeaux, Bât. B14, Allée Geoffroy Saint Hilaire, 33600, Pessac, France
| | - L Rouyer
- INSERM, BRIC, U 1312, University of Bordeaux, 33000, Bordeaux, France
| | - G Siegfried
- INSERM, BRIC, U 1312, University of Bordeaux, 33000, Bordeaux, France
- XenoFish, B2 Ouest, Allée Geoffroy St Hilaire CS50023, 33615, Pessac, France
| | - E Harté
- CNRS, LOMA, UMR 5798, University of Bordeaux, 33400, Talence, France
| | - F Bouvet
- CNRS, Bordeaux INP, CBMN, UMR 5248, University of Bordeaux, Bât. B14, Allée Geoffroy Saint Hilaire, 33600, Pessac, France
| | - L d'Agata
- CNRS, Bordeaux INP, CBMN, UMR 5248, University of Bordeaux, Bât. B14, Allée Geoffroy Saint Hilaire, 33600, Pessac, France
| | - E Darbo
- INSERM, BRIC, U 1312, University of Bordeaux, 33000, Bordeaux, France
| | - M Lefeuvre
- CNRS, Bordeaux INP, CBMN, UMR 5248, University of Bordeaux, Bât. B14, Allée Geoffroy Saint Hilaire, 33600, Pessac, France
| | - M A Derieppe
- Animalerie Mutualisée, Service Commun des Animaleries, University of Bordeaux, 33000, Bordeaux, France
| | - L Bouton
- INSERM, BRIC, U 1312, University of Bordeaux, 33000, Bordeaux, France
| | - M Mélane
- CNRS, LOMA, UMR 5798, University of Bordeaux, 33400, Talence, France
| | - D Chapeau
- CNRS, Bordeaux INP, CBMN, UMR 5248, University of Bordeaux, Bât. B14, Allée Geoffroy Saint Hilaire, 33600, Pessac, France
| | - J Martineau
- Animalerie Mutualisée, Service Commun des Animaleries, University of Bordeaux, 33000, Bordeaux, France
| | - V Prouzet-Mauleon
- INSERM, BRIC, U 1312, University of Bordeaux, 33000, Bordeaux, France
- CRISPRedit, TBMcore, UAR CNRS 3427, Inserm US 005, University of Bordeaux, Bordeaux, France
| | - S Tan
- CNRS, Bordeaux INP, CBMN, UMR 5248, University of Bordeaux, Bât. B14, Allée Geoffroy Saint Hilaire, 33600, Pessac, France
| | - W Souleyreau
- INSERM, BRIC, U 1312, University of Bordeaux, 33000, Bordeaux, France
| | - F Saltel
- INSERM, BRIC, U 1312, University of Bordeaux, 33000, Bordeaux, France
| | - F Argoul
- CNRS, LOMA, UMR 5798, University of Bordeaux, 33400, Talence, France
| | - A M Khatib
- INSERM, BRIC, U 1312, University of Bordeaux, 33000, Bordeaux, France
- XenoFish, B2 Ouest, Allée Geoffroy St Hilaire CS50023, 33615, Pessac, France
- Bergonié Institute, Bordeaux, France
| | - A R Brisson
- CNRS, Bordeaux INP, CBMN, UMR 5248, University of Bordeaux, Bât. B14, Allée Geoffroy Saint Hilaire, 33600, Pessac, France
| | - R Iggo
- INSERM, BRIC, U 1312, University of Bordeaux, 33000, Bordeaux, France
| | - A Bouter
- CNRS, Bordeaux INP, CBMN, UMR 5248, University of Bordeaux, Bât. B14, Allée Geoffroy Saint Hilaire, 33600, Pessac, France.
| |
Collapse
|
19
|
Chen SW, Blazeski A, Zhang S, Shelton SE, Offeddu GS, Kamm RD. Development of a perfusable, hierarchical microvasculature-on-a-chip model. LAB ON A CHIP 2023; 23:4552-4564. [PMID: 37771308 PMCID: PMC10563829 DOI: 10.1039/d3lc00512g] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/15/2023] [Indexed: 09/30/2023]
Abstract
Several methods have been developed for generating 3D, in vitro, organ-on-chip models of human vasculature to study vascular function, transport, and tissue engineering. However, many of these existing models lack the hierarchical nature of the arterial-to-capillary-to-venous architecture that is key to capturing a more comprehensive view of the human microvasculature. Here, we present a perfusable, multi-compartmental model that recapitulates the three microvascular compartments to assess various physiological properties such as vessel permeability, vasoconstriction dynamics, and circulating cell arrest and extravasation. Viscous finger patterning and passive pumping create the larger arterial and venular lumens, while the smaller diameter capillary bed vessels are generated through self-assembly. These compartments anastomose and form a perfusable, hierarchical system that portrays the directionality of blood flow through the microvasculature. The addition of collagen channels reduces the apparent permeability of the central capillary region, likely by reducing leakage from the side channels, enabling more accurate measurements of vascular permeability-an important motivation for this study. Furthermore, the model permits modulation of fluid flow and shear stress conditions throughout the system by using hydrostatic pressure heads to apply pressure differentials across either the arteriole or the capillary. This is a pertinent system for modeling circulating tumor or T cell dissemination and extravasation. Circulating cells were found to arrest in areas conducive to physical trapping or areas with the least amount of shear stress, consistent with hemodynamic or mechanical theories of metastasis. Overall, this model captures more features of human microvascular beds and is capable of testing a broad variety of hypotheses.
Collapse
Affiliation(s)
- Sophia W Chen
- Department of Biological Engineering, Massachusetts Institute of Technology, USA
| | - Adriana Blazeski
- Department of Biological Engineering, Massachusetts Institute of Technology, USA
- Department of Pathology, Brigham and Women's Hospital, USA
| | - Shun Zhang
- Department of Biological Engineering, Massachusetts Institute of Technology, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, USA
| | - Sarah E Shelton
- Department of Biological Engineering, Massachusetts Institute of Technology, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, USA
| | - Giovanni S Offeddu
- Department of Biological Engineering, Massachusetts Institute of Technology, USA
| | - Roger D Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, USA
| |
Collapse
|
20
|
Elanany MM, Mostafa D, Hamdy NM. Remodeled tumor immune microenvironment (TIME) parade via natural killer cells reprogramming in breast cancer. Life Sci 2023; 330:121997. [PMID: 37536617 DOI: 10.1016/j.lfs.2023.121997] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/20/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
Breast cancer (BC) is the main cause of cancer-related mortality among women globally. Despite substantial advances in the identification and management of primary tumors, traditional therapies including surgery, chemotherapy, and radiation cannot completely eliminate the danger of relapse and metastatic illness. Metastasis is controlled by microenvironmental and systemic mechanisms, including immunosurveillance. This led to the evolvement of immunotherapies that has gained much attention in the recent years for cancer treatment directed to the innate immune system. The long forgotten innate immune cells known as natural killer (NK) cells have emerged as novel targets for more effective therapeutics for BC. Normally, NK cells has the capacity to identify and eradicate tumor cells either directly or by releasing cytotoxic granules, chemokines and proinflammatory cytokines. Yet, NK cells are exposed to inhibitory signals by cancer cells, which causes them to become dysfunctional in the immunosuppressive tumor microenvironment (TME) in BC, supporting tumor escape and spread. Potential mechanisms of NK cell dysfunction in BC metastasis have been recently identified. Understanding these immunologic pathways driving BC metastasis will lead to improvements in the current immunotherapeutic strategies. In the current review, we highlight how BC evades immunosurveillance by rendering NK cells dysfunctional and we shed the light on novel NK cell- directed therapies.
Collapse
Affiliation(s)
- Mona M Elanany
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Ain Shams University, Abassia, 11566 Cairo, Egypt
| | - Dina Mostafa
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Ain Shams University, Abassia, 11566 Cairo, Egypt.
| | - Nadia M Hamdy
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Ain Shams University, Abassia, 11566 Cairo, Egypt.
| |
Collapse
|
21
|
Jung JI, Lee HS, Lee J, Kim EJ. Peanut sprout tea extract inhibits lung metastasis of 4T1 murine mammary carcinoma cells by suppressing the crosstalk between cancer cells and macrophages in BALB/c mice. Nutr Res Pract 2023; 17:917-933. [PMID: 37780222 PMCID: PMC10522819 DOI: 10.4162/nrp.2023.17.5.917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/09/2023] [Accepted: 06/07/2023] [Indexed: 10/03/2023] Open
Abstract
BACKGROUND/OBJECTIVES As peanuts germinate, the content of the components beneficial to health, such as resveratrol, increases within the peanut sprout. This study examined whether the ethanol extract of peanut sprout tea (PSTE) inhibits breast cancer growth and metastasis. MATERIALS/METHODS After orthotopically injecting 4T1 cells into BALB/c mice to induce breast cancer, 0, 30, or 60 mg/kg body weight/day of PSTE was administered orally. Angiogenesis-related protein expression in the tumors and the degree of metastasis were analyzed. 4T1 and RAW 264.7 cells were co-cultured, and reverse transcription polymerase chain reaction was performed to measure the crosstalk between breast cancer cells and macrophages. RESULTS PSTE reduced tumor growth and lung metastasis. In particular, PSTE decreased matrix metalloproteinase-9, platelet endothelial cell adhesion molecule-1, vascular endothelial growth factor-A, F4/80, CD11c, macrophage mannose receptor, macrophage colony-stimulating factor, and monocyte chemoattractant protein 1 expression in the tumors. Moreover, PSTE prevented 4T1 cell migration, invasion, and macrophage activity in RAW 264.7 cells. PSTE inhibited the crosstalk between 4T1 cells and RAW 264.7 cells and promoted the macrophage M1 subtype while inhibiting the M2 subtype. CONCLUSIONS These results suggest that PSTE blocks breast cancer growth and metastasis to the lungs. This may be because the PSTE treatment inhibits the crosstalk between mammary cancer cells and macrophages and inhibits the differentiation of macrophages into the M2 subtype.
Collapse
Affiliation(s)
- Jae In Jung
- Industry Coupled Cooperation Center for Bio Healthcare Materials, Hallym University, Chuncheon 24252, Korea
| | - Hyun Sook Lee
- Department of Food Science & Nutrition, Dongseo University, Busan 47011, Korea
| | - Jaehak Lee
- Department of Applied Animal Science, Kangwon National University, Chuncheon 24341, Korea
| | - Eun Ji Kim
- Industry Coupled Cooperation Center for Bio Healthcare Materials, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
22
|
Lahooti B, Akwii RG, Zahra FT, Sajib MS, Lamprou M, Alobaida A, Lionakis MS, Mattheolabakis G, Mikelis CM. Targeting endothelial permeability in the EPR effect. J Control Release 2023; 361:212-235. [PMID: 37517543 DOI: 10.1016/j.jconrel.2023.07.039] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 07/19/2023] [Accepted: 07/23/2023] [Indexed: 08/01/2023]
Abstract
The characteristics of the primary tumor blood vessels and the tumor microenvironment drive the enhanced permeability and retention (EPR) effect, which confers an advantage towards enhanced delivery of anti-cancer nanomedicine and has shown beneficial effects in preclinical models. Increased vascular permeability is a landmark feature of the tumor vessels and an important driver of the EPR. The main focus of this review is the endothelial regulation of vascular permeability. We discuss current challenges of targeting vascular permeability towards clinical translation and summarize the structural components and mechanisms of endothelial permeability, the principal mediators and signaling players, the targeted approaches that have been used and their outcomes to date. We also critically discuss the effects of the tumor-infiltrating immune cells, their interplay with the tumor vessels and the impact of immune responses on nanomedicine delivery, the impact of anti-angiogenic and tumor-stroma targeting approaches, and desirable nanoparticle design approaches for greater translational benefit.
Collapse
Affiliation(s)
- Behnaz Lahooti
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Racheal G Akwii
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Fatema Tuz Zahra
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Md Sanaullah Sajib
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Margarita Lamprou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras 26504, Greece
| | - Ahmed Alobaida
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, Ha'il 81442, Saudi Arabia
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - George Mattheolabakis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA.
| | - Constantinos M Mikelis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras 26504, Greece.
| |
Collapse
|
23
|
Tomassen T, Versleijen-Jonkers YMH, Hillebrandt-Roeffen MHS, Van Cleef PHJ, van Dalen T, Weidema ME, Desar IME, Flucke U, van Gorp JM. Prognostic Factors in Epithelioid Hemangioendothelioma: Analysis of a Nationwide Molecularly/Immunohistochemically Confirmed Cohort of 57 Cases. Cancers (Basel) 2023; 15:3304. [PMID: 37444414 DOI: 10.3390/cancers15133304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Epithelioid hemangioendothelioma (EHE) is an extremely rare vascular sarcoma with variable aggressive clinical behavior. In this retrospective study, we aimed to investigate prognostic factors based on clinicopathologic findings in a molecularly/immunohistochemically confirmed nationwide multicenter cohort of 57 EHE cases. Patients had unifocal disease (n = 29), multifocal disease (n = 5), lymph node metastasis (n = 8) and/or distant metastasis (n = 15) at the time of diagnosis. The overall survival rate was 71.4% at 1 year and 50.7% at 5 years. Survival did not correlate with sex, age or histopathological parameters. No survival differences were observed between multifocal and metastatic disease, suggesting that multifocality represents early metastases and treatment options are limited in comparison to unifocal disease. In unifocal tumors, survival could be predicted using the risk stratification model of Shibayama et al., dividing the cases into low- (n = 4), intermediate- (n = 15) and high- (n = 3) risk groups. No clinical or histopathological parameters were associated with progressive unifocal disease course. Lymph node metastases at the time of diagnosis occurred in 14.0% of the cases and were mainly associated with tumor localization in the head and neck area, proposing lymph node dissection. In conclusion, our results demonstrate the aggressive behavior of EHE, emphasize the prognostic value of a previously described risk stratification model and may provide new insights regarding tumor focality, therapeutic strategies and prognosis.
Collapse
Affiliation(s)
- Tess Tomassen
- Department of Pathology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Yvonne M H Versleijen-Jonkers
- Department of Medical Oncology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Melissa H S Hillebrandt-Roeffen
- Department of Medical Oncology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Patricia H J Van Cleef
- Department of Pathology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Thijs van Dalen
- Department of Surgery, Utrecht University Medical Center, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Marije E Weidema
- Department of Medical Oncology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Ingrid M E Desar
- Department of Medical Oncology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Uta Flucke
- Department of Pathology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
| | - Joost M van Gorp
- Department of Pathology, St. Antonius Hospital, Koekoekslaan 1, 3435 CM Nieuwegein, The Netherlands
| |
Collapse
|
24
|
Saito D, Tadokoro R, Nagasaka A, Yoshino D, Teramoto T, Mizumoto K, Funamoto K, Kidokoro H, Miyata T, Tamura K, Takahashi Y. Stiffness of primordial germ cells is required for their extravasation in avian embryos. iScience 2022; 25:105629. [PMID: 36465120 PMCID: PMC9713369 DOI: 10.1016/j.isci.2022.105629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/14/2022] [Accepted: 11/16/2022] [Indexed: 11/21/2022] Open
Abstract
Unlike mammals, primordial germ cells (PGCs) in avian early embryos exploit blood circulation to translocate to the somatic gonadal primordium, but how circulating PGCs undergo extravasation remains elusive. We demonstrate with single-cell level live-imaging analyses that the PGCs are arrested at a specific site in the capillary plexus, which is predominantly governed by occlusion at a narrow path in the vasculature. The occlusion is enabled by a heightened stiffness of the PGCs mediated by actin polymerization. Following the occlusion, PGCs reset their stiffness to soften in order to squeeze through the endothelial lining as they transmigrate. Our discovery also provides a model for the understanding of metastasizing cancer extravasation occurring mainly by occlusion.
Collapse
Affiliation(s)
- Daisuke Saito
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
- Department of Ecological Developmental Adaptability Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Fukuoka 819-0395, Japan
| | - Ryosuke Tadokoro
- Department of Zoology, Graduate School of Science, Kyoto University, Kyoto, Kyoto 606-8502, Japan
- Department of Bioscience, Okayama University of Science, Okayama, Okayama 700-0005, Japan
| | - Arata Nagasaka
- Division of Histology, Meikai University School of Dentistry, Sakado, Saitama 350-0283, Japan
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Daisuke Yoshino
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
- Institute of Engineering, Tokyo University of Agriculture and Technology, Koganei, Tokyo 184-8588, Japan
| | - Takayuki Teramoto
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Fukuoka 819-0395, Japan
| | - Kanta Mizumoto
- Department of Zoology, Graduate School of Science, Kyoto University, Kyoto, Kyoto 606-8502, Japan
| | - Kenichi Funamoto
- Institute of Fluid Science, Tohoku University, Sendai, Miyagi 980-8577, Japan
| | - Hinako Kidokoro
- Department of Ecological Developmental Adaptability Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
- Organization for Research Initiatives and Development, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
| | - Takaki Miyata
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Koji Tamura
- Department of Ecological Developmental Adaptability Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Yoshiko Takahashi
- Department of Zoology, Graduate School of Science, Kyoto University, Kyoto, Kyoto 606-8502, Japan
| |
Collapse
|
25
|
Overcoming challenges to enable targeting of metastatic breast cancer tumour microenvironment with nano-therapeutics: Current status and future perspectives. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
26
|
Wu Y, Zhou Y, Paul R, Qin X, Islam K, Liu Y. Adaptable Microfluidic Vessel-on-a-Chip Platform for Investigating Tumor Metastatic Transport in Bloodstream. Anal Chem 2022; 94:12159-12166. [PMID: 35998619 DOI: 10.1021/acs.analchem.2c02556] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cancer metastasis counts for 90% of cancer fatalities, and its development process is still a mystery. The dynamic process of tumor metastatic transport in the blood vessel is not well understood, in which some biomechanical factors, such as shear stress and various flow patterns, may have significant impacts. Here, we report a microfluidic vessel-on-a-chip platform for recapitulating several key metastatic steps of tumor cells in blood vessels on the same chip, including intravasation, circulating tumor cell (CTC) vascular adhesion, and extravasation. Due to its excellent adaptability, our system can reproduce various microenvironments to investigate the specific interactions between CTCs and blood vessels. On the basis of this platform, effects of important biomechanical factors on CTC adhesion such as vascular surface properties and vessel geometry-dependent hemodynamics were specifically inspected. We demonstrated that CTC adhesion is more likely to occur under certain mechano-physiological situations, such as vessels with vascular glycocalyx (VGCX) shedding and hemodynamic disturbances. Finally, computational models of both the fluidic dynamics in vessels and CTC adhesion were established based on the confocal scanned 3D images. The modeling results are believed to provide insights into exploring tumor metastasis progression and inspire new ideas for anticancer therapy development.
Collapse
Affiliation(s)
- Yue Wu
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Yuyuan Zhou
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Ratul Paul
- Department of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Xiaochen Qin
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Khayrul Islam
- Department of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Yaling Liu
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States.,Department of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| |
Collapse
|
27
|
Koszalka P, Kutryb-Zajac B, Mierzejewska P, Tomczyk M, Wietrzyk J, Serafin PK, Smolenski RT, Slominska EM. 4-Pyridone-3-carboxamide-1-β-D-ribonucleoside (4PYR)—A Novel Oncometabolite Modulating Cancer-Endothelial Interactions in Breast Cancer Metastasis. Int J Mol Sci 2022; 23:ijms23105774. [PMID: 35628582 PMCID: PMC9145394 DOI: 10.3390/ijms23105774] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/16/2022] [Accepted: 05/19/2022] [Indexed: 11/17/2022] Open
Abstract
The accumulation of specific metabolic intermediates is known to promote cancer progression. We analyzed the role of 4-pyridone-3-carboxamide-1-β-D-ribonucleoside (4PYR), a nucleotide metabolite that accumulates in the blood of cancer patients, using the 4T1 murine in vivo breast cancer model, and cultured cancer (4T1) and endothelial cells (ECs) for in vitro studies. In vivo studies demonstrated that 4PYR facilitated lung metastasis without affecting primary tumor growth. In vitro studies demonstrated that 4PYR affected extracellular adenine nucleotide metabolism and the intracellular energy status in ECs, shifting catabolite patterns toward the accumulation of extracellular inosine, and leading to the increased permeability of lung ECs. These changes prevailed over the direct effect of 4PYR on 4T1 cells that reduced their invasive potential through 4PYR-induced modulation of the CD73-adenosine axis. We conclude that 4PYR is an oncometabolite that affects later stages of the metastatic cascade by acting specifically through the regulation of EC permeability and metabolic controls of inflammation.
Collapse
Affiliation(s)
- Patrycja Koszalka
- Institute of Medical Biotechnology and Experimental Oncology, Intercollegiate Faculty of Biotechnology UG-MUG, Medical University of Gdansk, 80-210 Gdansk, Poland;
- Correspondence: (P.K.); (E.M.S.); Tel.: +48-58-349-1410 (P.K.); +48-58-349-1006 (E.M.S.)
| | - Barbara Kutryb-Zajac
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland; (B.K.-Z.); (P.M.); (M.T.); (R.T.S.)
| | - Paulina Mierzejewska
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland; (B.K.-Z.); (P.M.); (M.T.); (R.T.S.)
| | - Marta Tomczyk
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland; (B.K.-Z.); (P.M.); (M.T.); (R.T.S.)
| | - Joanna Wietrzyk
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland;
| | - Pawel K. Serafin
- Institute of Medical Biotechnology and Experimental Oncology, Intercollegiate Faculty of Biotechnology UG-MUG, Medical University of Gdansk, 80-210 Gdansk, Poland;
| | - Ryszard T. Smolenski
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland; (B.K.-Z.); (P.M.); (M.T.); (R.T.S.)
| | - Ewa M. Slominska
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland; (B.K.-Z.); (P.M.); (M.T.); (R.T.S.)
- Correspondence: (P.K.); (E.M.S.); Tel.: +48-58-349-1410 (P.K.); +48-58-349-1006 (E.M.S.)
| |
Collapse
|
28
|
Sarikaya I. Biology of Cancer and PET Imaging: Pictorial Review. J Nucl Med Technol 2022; 50:jnmt.121.263534. [PMID: 35440477 DOI: 10.2967/jnmt.121.263534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/10/2022] [Indexed: 11/16/2022] Open
Abstract
Development and spread of cancer is a multi-step and complex process which involves number of alterations, interactions and molecular networks. PET imaging is closely related with biology of cancer as it detects the cancer based on biological and pathological changes in tumor cells and tumor microenvironment. In this review article, biology of development and spread of cancer and role of PET imaging in Oncology was summarized and supported with various PET images demonstrating cancer spread patterns.
Collapse
|
29
|
Zhang H, Yu R, Zhang L, Wang R, Xiao L. Chemotherapy versus chemoradiotherapy for FIGO stages IB1 and IIA1 cervical squamous cancer patients with lymphovascular space invasion: a retrospective study. BMC Cancer 2022; 22:202. [PMID: 35193525 PMCID: PMC8864837 DOI: 10.1186/s12885-022-09309-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 02/17/2022] [Indexed: 11/20/2022] Open
Abstract
Purpose To evaluate the impact of different adjuvant therapy on IB1 and IIA1 stage cervical squamous cell cancer patients with lymphovascular space invasion. It also aimed to analyze the relationship between lymphovascular space invasion and other clinical pathological characteristics on IB1 and IIA1 stage cervical squamous cell cancer patients. Methods This retrospective observational study collected data of FIGO stages IB1 and IIA1 squamous cervical cancer patients at the First Affiliated Hospital of Chongqing Medical University between 2014 and 2018. A correlation analysis between lymphovascular space invasion and other clinical or pathological factors was conducted. Prognosis analysis of patients with lymphovascular space invasion were performed to assess associations between clinical-pathological characteristics and survival. Results A total of 357 women were identified including 110 (30.8%) with lymphovascular space invasion, 247 (69.2%) without lymphovascular space invasion. Both middle 1/3 cervical stromal invasion (p = 0.000) and deep 1/3 cervical stromal invasion (p = 0.000) were independently associated with lymphovascular space invasion. Among lymphovascular space involved women, tumor differentiation (P = 0.001) and postoperative therapy (P = 0.036) had a significant influence on disease recurrence. Multivariate analysis showed that lymph node metastasis (P = 0.017), poorer tumor differentiation (P = 0.036) and postoperative chemotherapy alone (P = 0.021) can increase the risk of tumor relapse. Conclusion Our study suggested that the presence of deep stromal invasion independently increases the risk of lymphovascular space invasion. Compared with chemotherapy, chemoradiotherapy seems to improve progression-free survival in squamous cervical cancer patients with lymphovascular space invasion.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Rao Yu
- Department of Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lan Zhang
- Department of Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Rong Wang
- Department of Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lin Xiao
- Department of Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
30
|
Hou S, Wang J, Li W, Hao X, Hang Q. Roles of Integrins in Gastrointestinal Cancer Metastasis. Front Mol Biosci 2021; 8:708779. [PMID: 34869579 PMCID: PMC8634653 DOI: 10.3389/fmolb.2021.708779] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
Integrins are a large family of heterodimeric transmembrane receptors which mediate cell adhesion and transmit signals to the cell interior. The mechanistic roles of integrins have long been an enigma in cancer, given its complexity in regulating different cellular behaviors. Recently, however, increasing research is providing new insights into its function and the underlying mechanisms, which collectively include the influences of altered integrin expression on the aberrant signaling pathways and cancer progression. Many studies have also demonstrated the potentiality of integrins as therapeutic targets in cancer treatment. In this review, we have summarized these recent reports and put a particular emphasis on the dysregulated expression of integrins and how they regulate related signaling pathways to facilitate the metastatic progression of gastrointestinal cancer, including gastric cancer (GC) and colorectal cancer (CRC), which will address the crucial roles of integrins in gastrointestinal cancer.
Collapse
Affiliation(s)
- Sicong Hou
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Jiaxin Wang
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Wenqian Li
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Xin Hao
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Qinglei Hang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
31
|
Chugh V, Vijaya Krishna K, Pandit A. Cell Membrane-Coated Mimics: A Methodological Approach for Fabrication, Characterization for Therapeutic Applications, and Challenges for Clinical Translation. ACS NANO 2021; 15:17080-17123. [PMID: 34699181 PMCID: PMC8613911 DOI: 10.1021/acsnano.1c03800] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 10/13/2021] [Indexed: 05/04/2023]
Abstract
Cell membrane-coated (CMC) mimics are micro/nanosystems that combine an isolated cell membrane and a template of choice to mimic the functions of a cell. The design exploits its physicochemical and biological properties for therapeutic applications. The mimics demonstrate excellent biological compatibility, enhanced biointerfacing capabilities, physical, chemical, and biological tunability, ability to retain cellular properties, immune escape, prolonged circulation time, and protect the encapsulated drug from degradation and active targeting. These properties and the ease of adapting them for personalized clinical medicine have generated a significant research interest over the past decade. This review presents a detailed overview of the recent advances in the development of cell membrane-coated (CMC) mimics. The primary focus is to collate and discuss components, fabrication methodologies, and the significance of physiochemical and biological characterization techniques for validating a CMC mimic. We present a critical analysis of the two main components of CMC mimics: the template and the cell membrane and mapped their use in therapeutic scenarios. In addition, we have emphasized on the challenges associated with CMC mimics in their clinical translation. Overall, this review is an up to date toolbox that researchers can benefit from while designing and characterizing CMC mimics.
Collapse
Affiliation(s)
| | | | - Abhay Pandit
- CÚRAM, SFI Research
Centre for Medical Devices, National University
of Ireland Galway, Galway H91 W2TY, Ireland
| |
Collapse
|
32
|
Colin F, Gensbittel V, Goetz JG. Biomechanics: a driving force behind metastatic progression. C R Biol 2021; 344:249-262. [DOI: 10.5802/crbiol.62] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 10/04/2021] [Indexed: 11/24/2022]
|
33
|
Middleton JD, Sivakumar S, Hai T. Chemotherapy-Induced Changes in the Lung Microenvironment: The Role of MMP-2 in Facilitating Intravascular Arrest of Breast Cancer Cells. Int J Mol Sci 2021; 22:10280. [PMID: 34638621 PMCID: PMC8508901 DOI: 10.3390/ijms221910280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/14/2021] [Accepted: 09/17/2021] [Indexed: 11/18/2022] Open
Abstract
Previously, we showed that mice treated with cyclophosphamide (CTX) 4 days before intravenous injection of breast cancer cells had more cancer cells in the lung at 3 h after cancer injection than control counterparts without CTX. At 4 days after its injection, CTX is already excreted from the mice, allowing this pre-treatment design to reveal how CTX may modify the lung environment to indirectly affect cancer cells. In this study, we tested the hypothesis that the increase in cancer cell abundance at 3 h by CTX is due to an increase in the adhesiveness of vascular wall for cancer cells. Our data from protein array analysis and inhibition approach combined with in vitro and in vivo assays support the following two-prong mechanism. (1) CTX increases vascular permeability, resulting in the exposure of the basement membrane (BM). (2) CTX increases the level of matrix metalloproteinase-2 (MMP-2) in mouse serum, which remodels the BM and is functionally important for CTX to increase cancer abundance at this early stage. The combined effect of these two processes is the increased accessibility of critical protein domains in the BM, resulting in higher vascular adhesiveness for cancer cells to adhere. The critical protein domains in the vascular microenvironment are RGD and YISGR domains, whose known binding partners on cancer cells are integrin dimers and laminin receptor, respectively.
Collapse
Affiliation(s)
- Justin D. Middleton
- Department of Biological Chemistry and Pharmacology, College of Medicine, Ohio State University, Columbus, OH 43210, USA; (J.D.M.); (S.S.)
- Molecular, Cellular, and Developmental Biology Program, Ohio State University, Columbus, OH 43210, USA
| | - Subhakeertana Sivakumar
- Department of Biological Chemistry and Pharmacology, College of Medicine, Ohio State University, Columbus, OH 43210, USA; (J.D.M.); (S.S.)
| | - Tsonwin Hai
- Department of Biological Chemistry and Pharmacology, College of Medicine, Ohio State University, Columbus, OH 43210, USA; (J.D.M.); (S.S.)
- Molecular, Cellular, and Developmental Biology Program, Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
34
|
Allen SC, Widman JA, Datta A, Suggs LJ. Dynamic extracellular matrix stiffening induces a phenotypic transformation and a migratory shift in epithelial cells. Integr Biol (Camb) 2021; 12:161-174. [PMID: 32472133 DOI: 10.1093/intbio/zyaa012] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 12/10/2019] [Accepted: 05/01/2020] [Indexed: 12/21/2022]
Abstract
Soft tissue tumors, including breast cancer, become stiffer throughout disease progression. This increase in stiffness has been shown to correlate to malignant phenotype and epithelial-to-mesenchymal transition (EMT) in vitro. Unlike current models, utilizing static increases in matrix stiffness, our group has previously created a system that allows for dynamic stiffening of an alginate-matrigel composite hydrogel to mirror the native dynamic process. Here, we utilize this system to evaluate the role of matrix stiffness on EMT and metastasis both in vitro and in vivo. Epithelial cells were seen to lose normal morphology and become protrusive and migratory after stiffening. This shift corresponded to a loss of epithelial markers and gain of mesenchymal markers in both the cell clusters and migrated cells. Furthermore, stiffening in a murine model reduced tumor burden and increased migratory behavior prior to tumor formation. Inhibition of FAK and PI3K in vitro abrogated the morphologic and migratory transformation of epithelial cell clusters. This work demonstrates the key role extracellular matrix stiffening has in tumor progression through integrin signaling and, in particular, its ability to drive EMT-related changes and metastasis.
Collapse
Affiliation(s)
- Shane C Allen
- Department of Biomedical Engineering, The University of Texas, Austin, TX, USA
| | - Jessica A Widman
- Department of Biomedical Engineering, The University of Texas, Austin, TX, USA
| | - Anisha Datta
- Department of Biomedical Engineering, The University of Texas, Austin, TX, USA
| | - Laura J Suggs
- Department of Biomedical Engineering, The University of Texas, Austin, TX, USA
| |
Collapse
|
35
|
Kwan YP, Teo MHY, Lim JCW, Tan MS, Rosellinny G, Wahli W, Wang X. LRG1 Promotes Metastatic Dissemination of Melanoma through Regulating EGFR/STAT3 Signalling. Cancers (Basel) 2021; 13:3279. [PMID: 34208965 PMCID: PMC8269286 DOI: 10.3390/cancers13133279] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/19/2021] [Accepted: 06/23/2021] [Indexed: 12/17/2022] Open
Abstract
Although less common, melanoma is the deadliest form of skin cancer largely due to its highly metastatic nature. Currently, there are limited treatment options for metastatic melanoma and many of them could cause serious side effects. A better understanding of the molecular mechanisms underlying the complex disease pathophysiology of metastatic melanoma may lead to the identification of novel therapeutic targets and facilitate the development of targeted therapeutics. In this study, we investigated the role of leucine-rich α-2-glycoprotein 1 (LRG1) in melanoma development and progression. We first established the association between LRG1 and melanoma in both human patient biopsies and mouse melanoma cell lines and revealed a significant induction of LRG1 expression in metastatic melanoma cells. We then showed no change in tumour cell growth, proliferation, and angiogenesis in the absence of the host Lrg1. On the other hand, there was reduced melanoma cell metastasis to the lungs in Lrg1-deficient mice. This observation was supported by the promoting effect of LRG1 in melanoma cell migration, invasion, and adhesion. Mechanistically, LRG1 mediates melanoma cell invasiveness in an EGFR/STAT3-dependent manner. Taken together, our studies provided compelling evidence that LRG1 is required for melanoma metastasis but not growth. Targeting LRG1 may offer an alternative strategy to control malignant melanoma.
Collapse
Affiliation(s)
- Yuet Ping Kwan
- Centre for Vision Research, Duke NUS Medical School, 8 College Road, Singapore 169857, Singapore; (Y.P.K.); (M.H.Y.T.); (G.R.)
- Singapore Eye Research Institute (SERI) The Academia, 20 College Road, Level 6 Discovery Tower, Singapore 169856, Singapore
| | - Melissa Hui Yen Teo
- Centre for Vision Research, Duke NUS Medical School, 8 College Road, Singapore 169857, Singapore; (Y.P.K.); (M.H.Y.T.); (G.R.)
- Singapore Eye Research Institute (SERI) The Academia, 20 College Road, Level 6 Discovery Tower, Singapore 169856, Singapore
| | - Jonathan Chee Woei Lim
- Pharmacotherapeutics Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
| | - Michelle Siying Tan
- Department of Surgery, Yong Yoo Lin School of Medicine, National University of Singapore, MD6, 14 Medical Drive, Singapore 117599, Singapore;
| | - Graciella Rosellinny
- Centre for Vision Research, Duke NUS Medical School, 8 College Road, Singapore 169857, Singapore; (Y.P.K.); (M.H.Y.T.); (G.R.)
- Singapore Eye Research Institute (SERI) The Academia, 20 College Road, Level 6 Discovery Tower, Singapore 169856, Singapore
| | - Walter Wahli
- Center for Integrative Genomics, Université de Lausanne, Le Génopode, CH-1015 Lausanne, Switzerland;
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11 Mandalay Road, Singapore 308232, Singapore
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR 1331, UPS, Université de Toulouse, F-31027 Toulouse, France
| | - Xiaomeng Wang
- Centre for Vision Research, Duke NUS Medical School, 8 College Road, Singapore 169857, Singapore; (Y.P.K.); (M.H.Y.T.); (G.R.)
- Singapore Eye Research Institute (SERI) The Academia, 20 College Road, Level 6 Discovery Tower, Singapore 169856, Singapore
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Proteos, 61 Biopolis Dr, Singapore 138673, Singapore
| |
Collapse
|
36
|
Circulating Tumor Cells from Enumeration to Analysis: Current Challenges and Future Opportunities. Cancers (Basel) 2021; 13:cancers13112723. [PMID: 34072844 PMCID: PMC8198976 DOI: 10.3390/cancers13112723] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 05/25/2021] [Indexed: 01/19/2023] Open
Abstract
Simple Summary With estimated numbers of 1–10 per mL of blood, circulating tumor cells (CTCs) are extremely rare compared to white (a few million) or red (billions) blood cells. Given their critical role in metastasis, CTCs have enormous potential as a biomarker for cancer diagnosis, prognosis, and monitoring of treatment response. There are now efforts to characterize CTCs more precisely through molecular and functional analysis, expanding the CTC effort from one of diagnosis and prognosis to now include the use of CTCs to specifically target cancers and discover therapeutic solutions, establishing CTCs as critical in precision medicine. This article summarizes current knowledge about CTC isolation technologies and discusses the translational benefits of different types of downstream analysis approaches, including single-CTC analysis, ex vivo expansion of CTCs, and characterization of CTC-associated cells. Abstract Circulating tumor cells (CTCs) have been recognized as a major contributor to distant metastasis. Their unique role as metastatic seeds renders them a potential marker in the circulation for early cancer diagnosis and prognosis as well as monitoring of therapeutic response. In the past decade, researchers mainly focused on the development of isolation techniques for improving the recovery rate and purity of CTCs. These developed techniques have significantly increased the detection sensitivity and enumeration accuracy of CTCs. Currently, significant efforts have been made toward comprehensive molecular characterization, ex vivo expansion of CTCs, and understanding the interactions between CTCs and their associated cells (e.g., immune cells and stromal cells) in the circulation. In this review, we briefly summarize existing CTC isolation technologies and specifically focus on advances in downstream analysis of CTCs and their potential applications in precision medicine. We also discuss the current challenges and future opportunities in their clinical utilization.
Collapse
|
37
|
Lee G, Yoon S, Ahn B, Kim HR, Jang SJ, Hwang HS. Blood Vessel Invasion Predicts Postoperative Survival Outcomes and Systemic Recurrence Regardless of Location or Blood Vessel Type in Patients with Lung Adenocarcinoma. Ann Surg Oncol 2021; 28:7279-7290. [PMID: 34041629 DOI: 10.1245/s10434-021-10122-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/18/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Presence of blood vessel invasion (BVI) is one of the prognostic indicators for lung cancer patients with surgical resection. However, prognostic roles of the location and the type of the involved blood vessel have not been fully evaluated yet. PATIENTS AND METHODS We retrieved the data of 217 cases of surgically resected lung adenocarcinoma from Asan Medical Center. Clinicopathologic features, including BVI, were reassessed. The location (tumor center and/or periphery) and involved blood vessel types (large and/or small vessels; arteries and/or veins) of BVI were separately examined on standard hematoxylin-eosin slides and confirmed by van Gieson elastic staining. RESULTS BVI was identified in 35% of cases (76/217), with the tumor center (intratumoral) as the location in more than half of the cases (42/76, 55.3%). The presence of BVI was significantly associated with higher pathologic stage, increased size of invasive components, frequent pleural invasion, lymphatic permeation, and spread through alveolar spaces. BVI was significantly associated with poor overall survival (OS) and recurrence-free survival (RFS) both in univariate and multivariate survival analyses [for OS, hazard ratio (HR) 1.92, 95% confidence interval (CI) 1.06-3.48, P = 0.031; for RFS, HR 2.65, 95% CI 1.64-4.28; P < 0.001]. BVI subgroups, according to location and type of the involved blood vessels, invariably displayed significantly poor RFS; however, the results for OS varied. CONCLUSION Regardless of their location or blood vessel type, presence of BVI is a useful predictor for postoperative survival outcomes, which should be carefully evaluated on pathologic examination.
Collapse
Affiliation(s)
- Goeun Lee
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Shinkyo Yoon
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Bokyung Ahn
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyeong-Ryul Kim
- Department of Chest surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Se Jin Jang
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hee Sang Hwang
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
38
|
Jin Y, Liu W, Wang F, Wang M, Xu K, Yang A, Wang C, Zhang L, Zhang F, Li M. Tissue factor potentiates adherence of breast cancer cells to human umbilical vein endothelial cells under static and flow conditions. Cell Adh Migr 2021; 15:74-83. [PMID: 33734001 PMCID: PMC7993123 DOI: 10.1080/19336918.2021.1898709] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Tissue factor (TF) has been extensively studied for tumor metastasis, but its role in mediating cancer cell adhesion to vasculature remains unknown. This study aimed to measure the ability of TF to mediate the adhesion of breast cancer cells to human umbilical vein endothelial cells (HUVECs). MDA-MB-231 cells expressed the highest TF level and adhered more to HUVECs under static and flow conditions, a neutralizing TF antibody abolished the enhanced adhesion of MDA-MB-231 cells to HUVECs. Recombinant human soluble TF (rTF) bonded β1integrin on HUVECs surfaces, β1 or α3integrin antibody combined with TF antibody abolished more cell-cell adhesion. These data suggested that TF mediated adhesion of breast cancer cells to endothelial cells may rely on β1integrin on HUVECs surfaces.
Collapse
Affiliation(s)
- Yanling Jin
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Wei Liu
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Fengxia Wang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Min Wang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Kai Xu
- First Affiliated Clinical Hospital, Lanzhou University, Lanzhou, Gansu, China
| | - Aijun Yang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Chenyu Wang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Lihan Zhang
- Department of Integrated Traditional Chinese and Western Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fangfang Zhang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Min Li
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Gansu Provincial Key Laboratory of Preclinical Study for New Drug Development, Lanzhou University, Lanzhou, China
| |
Collapse
|
39
|
Muscarella AM, Aguirre S, Hao X, Waldvogel SM, Zhang XHF. Exploiting bone niches: progression of disseminated tumor cells to metastasis. J Clin Invest 2021; 131:143764. [PMID: 33720051 PMCID: PMC7954594 DOI: 10.1172/jci143764] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Many solid cancers metastasize to the bone and bone marrow (BM). This process may occur even before the diagnosis of primary tumors, as evidenced by the discovery of disseminated tumor cells (DTCs) in patients without occult malignancies. The cellular fates and metastatic progression of DTCs are determined by complicated interactions between cancer cells and BM niches. Not surprisingly, these niches also play important roles in normal biology, including homeostasis and turnover of skeletal and hematopoiesis systems. In this Review, we summarize recent findings on functions of BM niches in bone metastasis (BoMet), particularly during the early stage of colonization. In light of the rich knowledge of hematopoiesis and osteogenesis, we highlight how DTCs may progress into overt BoMet by taking advantage of niche cells and their activities in tissue turnover, especially those related to immunomodulation and bone repair.
Collapse
Affiliation(s)
- Aaron M. Muscarella
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, Texas, USA
| | - Sergio Aguirre
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, Texas, USA
| | - Xiaoxin Hao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Sarah M. Waldvogel
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas, USA
| | - Xiang H.-F. Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- McNair Medical Institute, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
40
|
Sigdel I, Gupta N, Faizee F, Khare VM, Tiwari AK, Tang Y. Biomimetic Microfluidic Platforms for the Assessment of Breast Cancer Metastasis. Front Bioeng Biotechnol 2021; 9:633671. [PMID: 33777909 PMCID: PMC7992012 DOI: 10.3389/fbioe.2021.633671] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 02/05/2021] [Indexed: 12/27/2022] Open
Abstract
Of around half a million women dying of breast cancer each year, more than 90% die due to metastasis. Models necessary to understand the metastatic process, particularly breast cancer cell extravasation and colonization, are currently limited and urgently needed to develop therapeutic interventions necessary to prevent breast cancer metastasis. Microfluidic approaches aim to reconstitute functional units of organs that cannot be modeled easily in traditional cell culture or animal studies by reproducing vascular networks and parenchyma on a chip in a three-dimensional, physiologically relevant in vitro system. In recent years, microfluidics models utilizing innovative biomaterials and micro-engineering technologies have shown great potential in our effort of mechanistic understanding of the breast cancer metastasis cascade by providing 3D constructs that can mimic in vivo cellular microenvironment and the ability to visualize and monitor cellular interactions in real-time. In this review, we will provide readers with a detailed discussion on the application of the most up-to-date, state-of-the-art microfluidics-based breast cancer models, with a special focus on their application in the engineering approaches to recapitulate the metastasis process, including invasion, intravasation, extravasation, breast cancer metastasis organotropism, and metastasis niche formation.
Collapse
Affiliation(s)
- Indira Sigdel
- Biofluidics Laboratory, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| | - Niraj Gupta
- Biofluidics Laboratory, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| | - Fairuz Faizee
- Biofluidics Laboratory, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| | - Vishwa M Khare
- Eurofins Lancaster Laboratories, Philadelphia, PA, United States
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH, United States
| | - Yuan Tang
- Biofluidics Laboratory, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| |
Collapse
|
41
|
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: 8.0] [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
|
42
|
Tuffour A, Kosiba AA, Zhang Y, Peprah FA, Gu J, Shi H. Role of the calcium-sensing receptor (CaSR) in cancer metastasis to bone: Identifying a potential therapeutic target. Biochim Biophys Acta Rev Cancer 2021; 1875:188528. [PMID: 33640382 DOI: 10.1016/j.bbcan.2021.188528] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 02/16/2021] [Accepted: 02/20/2021] [Indexed: 12/20/2022]
Abstract
Cancer is a major cause of morbidity and mortality worldwide due to its ability to evade immune surveillance and metastasize from its origin to a secondary point of contact. Though several treatment techniques have been developed to suppress or manage cancer spread, a strategy for total control over the disease continues to evade researchers. In considering ways to control or prevent cancer from metastasizing to the bone, we analyze the impact of the calcium-sensing receptor (CaSR), whose primary role is to maintain calcium (Ca2+) homeostasis in cellular and systemic physiological processes. CaSR is a pleiotropic receptor capable of enhancing the proliferation of some cancers such as breast, lung, prostate and kidney cancers at its primary site(s) and stimulating bone metastasis, while exerting a suppressive effect in others such as colon cancer. The activity of CaSR not only increases cancer cell proliferation, migration and suppression of apoptosis in the organs indicated, but also increases the secretion of parathyroid hormone-related protein (PTHrP) and epiregulin, which induce osteolytic activity and osteoblastic suppression. In addition, released cytokines and Ca2+ from bone resorption are critical factors that further promote cancer proliferation. In this review, we seek to highlight previous viewpoints on CaSR, discuss its role in a new context, and consider its potential clinical application in cancer treatment.
Collapse
Affiliation(s)
- Alex Tuffour
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| | | | - Yao Zhang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Frank Addai Peprah
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Jie Gu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Haifeng Shi
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| |
Collapse
|
43
|
The role of long noncoding RNAs in regulating invasion and metastasis of malignant tumors. Anticancer Drugs 2021; 31:319-325. [PMID: 32011368 DOI: 10.1097/cad.0000000000000899] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Long noncoding RNAs (lncRNAs) are a group of non-protein-coding transcripts exceeding 200 nucleotides in length, which are emerging as key players in various fundamental biological processes. Furthermore, it is increasingly recognized that mutation and dysregulation of lncRNAs contribute importantly to a variety of human diseases, particularly human cancers. Previous studies have revealed that altered lncRNAs have a close association with tumorigenesis, metastasis, prognosis and diagnosis of cancers. The present review aims to exhibit a brief overview of the associated reports of lncRNAs in cancers, including colorectal cancer, gastric cancer, lung adenocarcinoma, nasopharyngeal carcinoma, cervical cancer and esophageal cancer. Altogether, we argue that lncRNAs have potential as new biomarkers in cancer prognosis and diagnosis, and as promising therapeutic targets for the prevention and treatment of human cancers.
Collapse
|
44
|
Lolo FN, Jiménez-Jiménez V, Sánchez-Álvarez M, Del Pozo MÁ. Tumor-stroma biomechanical crosstalk: a perspective on the role of caveolin-1 in tumor progression. Cancer Metastasis Rev 2021; 39:485-503. [PMID: 32514892 DOI: 10.1007/s10555-020-09900-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tumor stiffening is a hallmark of malignancy that actively drives tumor progression and aggressiveness. Recent research has shed light onto several molecular underpinnings of this biomechanical process, which has a reciprocal crosstalk between tumor cells, stromal fibroblasts, and extracellular matrix remodeling at its core. This dynamic communication shapes the tumor microenvironment; significantly determines disease features including therapeutic resistance, relapse, or metastasis; and potentially holds the key for novel antitumor strategies. Caveolae and their components emerge as integrators of different aspects of cell function, mechanotransduction, and ECM-cell interaction. Here, we review our current knowledge on the several pivotal roles of the essential caveolar component caveolin-1 in this multidirectional biomechanical crosstalk and highlight standing questions in the field.
Collapse
Affiliation(s)
- Fidel Nicolás Lolo
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Víctor Jiménez-Jiménez
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Miguel Sánchez-Álvarez
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Miguel Ángel Del Pozo
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
45
|
Li B, Xu H, He C, Zou W, Tu Y. Lidocaine prevents breast cancer growth by targeting neuronatin to inhibit nerve fibers formation. J Toxicol Sci 2021; 46:329-339. [PMID: 34193770 DOI: 10.2131/jts.46.329] [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] [Indexed: 12/24/2022]
Abstract
Lidocaine has been shown to inhibit the invasion and metastasis of breast cancer, but the mechanism still remains unclear. This study explored the relationship between lidocaine and circulating seeding of breast cancer cells from the perspective of nerve fiber formation. The cell lines MDA-MB-231 and 4T1 were subcutaneously inoculated in mice to simulate the tumor self-seeding by circulating cancer cells. Lidocaine was used to treat these mice and tumor growth was observed. Silver staining was performed to observe the distribution of nerve fibers in tumor-bearing tissues, and immunohistochemical analysis was performed to observe the expression levels of nerve-related proteins. The results showed that lidocaine treatment effectively inhibited tumor growth and nerve fiber formation, and down-regulated the expression levels of protein gene product 9.5, neurofilament, nerve growth factor (NGF), and neuronatin (Nnat). Overexpression NGF and Nnat both could reverse the therapeutic effects of lidocaine. These results suggest that the effect of lidocaine on inhibiting breast cancer invasion and metastasis may be achieved by targeting Nnat, regulating the production of NGFs in cancer cells, and subsequently inhibiting the formation of nerve fibers.
Collapse
Affiliation(s)
- Bingda Li
- Department of Anesthesiology, Jiangxi Cancer Hospital of Nanchang University, China
| | - Hao Xu
- Department of Pediatrics, Wuhan NO.1 Hospital, China
| | - Chongwu He
- Department of Breast Surgery, Jiangxi Cancer Hospital of Nanchang University, China
| | - Wenxiong Zou
- Department of Emergency, Ji'an Central People's Hospital, China
| | - Yun Tu
- Department of Oncology, Jiangxi Cancer Hospital of Nanchang University, China
| |
Collapse
|
46
|
Stock C. Circulating Tumor Cells: Does Ion Transport Contribute to Intravascular Survival, Adhesion, Extravasation, and Metastatic Organotropism? Rev Physiol Biochem Pharmacol 2021; 182:139-175. [DOI: 10.1007/112_2021_68] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
47
|
Mo Z, Cheong JYA, Xiang L, Le MTN, Grimson A, Zhang DX. Extracellular vesicle-associated organotropic metastasis. Cell Prolif 2021; 54:e12948. [PMID: 33145869 PMCID: PMC7791170 DOI: 10.1111/cpr.12948] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/28/2020] [Accepted: 10/14/2020] [Indexed: 12/11/2022] Open
Abstract
Metastasis refers to the progressive dissemination of primary tumour cells and their colonization of other tissues and is associated with most cancer-related mortalities. The disproportional and systematic distribution pattern of distant metastasis in different cancers has been well documented, as is termed metastatic organotropism, a process orchestrated by a combination of anatomical, pathophysiological, genetic and biochemical factors. Extracellular vesicles (EVs), nanosized cell-derived membrane-bound particles known to mediate intercellular communication, are now considered crucial in organ-specific metastasis. Here, we review and summarize recent findings regarding EV-associated organotropic metastasis as well as some of the general mechanisms by which EVs contribute to this important process in cancer and provide a future perspective on this emerging topic. We highlight studies that demonstrate a role of tumour-derived EVs in organotropic metastasis via pre-metastatic niche modulation. The bioactive cargo carried by EVs is of diagnostic and prognostic values, and counteracting the functions of such EVs may be a novel therapeutic strategy targeting metastasis. Further investigations are warranted to better understand the functions and mechanisms of EVs in organotropic metastasis and accelerate the relevant clinical translation.
Collapse
Affiliation(s)
- Zhenzhen Mo
- Department of PaediatricsPeople's Hospital of Guangxi Zhuang Autonomous RegionNanningChina
| | - Jia Yang Alex Cheong
- Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Lirong Xiang
- Department of PaediatricsPeople's Hospital of Guangxi Zhuang Autonomous RegionNanningChina
| | - Minh T. N. Le
- Institute for Digital Medicine and Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Andrew Grimson
- Department of Molecular Biology and GeneticsCornell UniversityIthacaNYUSA
| | - Daniel Xin Zhang
- Department of Biomedical SciencesJockey Club College of Veterinary Medicine and Life SciencesCity University of Hong KongKowloonHong Kong SAR
- Department of Molecular Biology and GeneticsCornell UniversityIthacaNYUSA
| |
Collapse
|
48
|
Riggio AI, Varley KE, Welm AL. The lingering mysteries of metastatic recurrence in breast cancer. Br J Cancer 2021; 124:13-26. [PMID: 33239679 PMCID: PMC7782773 DOI: 10.1038/s41416-020-01161-4] [Citation(s) in RCA: 306] [Impact Index Per Article: 76.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 02/07/2023] Open
Abstract
Despite being the hallmark of cancer that is responsible for the highest number of deaths, very little is known about the biology of metastasis. Metastatic disease typically manifests after a protracted period of undetectable disease following surgery or systemic therapy, owing to relapse or recurrence. In the case of breast cancer, metastatic relapse can occur months to decades after initial diagnosis and treatment. In this review, we provide an overview of the known key factors that influence metastatic recurrence, with the goal of highlighting the critical unanswered questions that still need to be addressed to make a difference in the mortality of breast cancer patients.
Collapse
Affiliation(s)
- Alessandra I Riggio
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Katherine E Varley
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Alana L Welm
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
49
|
Lu X, Tan S, Wu M, Ju H, Liang X, Li P. Evaluation of a new magnetic bead as an integrated platform for systematic CTC recognition, capture and clinical analysis. Colloids Surf B Biointerfaces 2020; 199:111542. [PMID: 33373845 DOI: 10.1016/j.colsurfb.2020.111542] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 11/11/2020] [Accepted: 12/12/2020] [Indexed: 12/28/2022]
Abstract
A novel form of magnetic bead, namely antibody-coated magnetic lipid nano-vehicle (AMLV), was synthesized by embedding Fe3O4 nanoparticles into an amphiphilic antibody-modified liposome as a high-performance circulating tumor cell (CTC) hunter. The CTC capture performance of AMLV was validated based on an enlarged patient sample (including 318 colorectal, 78 breast, 77 lung and 55 liver cancer patients) with high detection rate. The preliminary comparison with Cellsearch was also conducted, indicating that the cell membrane-semblance AMLVEpCAM showed higher capture performance for different kinds of EpCAM-expressed circulating tumor cells in the peripheral blood (4.4 ± 1.2-fold for AMLVEpCAM vs CellsearchTM, n=5, P<0.001). Moreover, the AMLVEpCAM-isolated CTCs could be used as a functional material to provide various clinical information for tumor patients and work as an alternative of tumor tissue to conduct gene analysis after conventional PCR amplification.
Collapse
Affiliation(s)
- Xinmiao Lu
- Department of Nuclear Medicine Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, PR China
| | - Sheng Tan
- Department of Cardiothoracic Surgery, The Affiliated Hospital of XuZhou Medical University, Xuzhou, PR China
| | - Muyu Wu
- Department of Nuclear Medicine Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, PR China
| | - Huijun Ju
- Department of Nuclear Medicine Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, PR China
| | - Xiaofei Liang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln2200, XieTu Rd, Shanghai 200032, PR China.
| | - Peiyong Li
- Department of Nuclear Medicine Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, PR China.
| |
Collapse
|
50
|
Kim J, Lee C, Kim I, Ro J, Kim J, Min Y, Park J, Sunkara V, Park YS, Michael I, Kim YA, Lee HJ, Cho YK. Three-Dimensional Human Liver-Chip Emulating Premetastatic Niche Formation by Breast Cancer-Derived Extracellular Vesicles. ACS NANO 2020; 14:14971-14988. [PMID: 32880442 DOI: 10.1021/acsnano.0c04778] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The liver is one of the most common sites of breast cancer metastasis and is associated with high lethality. Although the interaction between tumor cells and their microenvironment at metastatic sites has been recognized as a key regulator of tumor progression, the underlying mechanism is not fully elucidated. Here, we describe a three-dimensional (3D) microfluidic human liver-on-a-chip (liver-chip) that emulates the formation of a premetastatic niche to investigate the roles of breast cancer-derived extracellular vesicles (EVs) in liver metastasis. We demonstrate that breast cancer-derived EVs activate liver sinusoidal endothelial cells (LSECs) in the liver-chip, inducing endothelial to mesenchymal transition and destruction of vessel barriers. In addition, we show that transforming growth factor β1 (TGFβ1) in breast cancer-derived EVs upregulates fibronectin, an adhesive extracellular matrix protein, on LSECs, which facilitates the adhesion of breast cancer cells to the liver microenvironment. Furthermore, we observed that EVs isolated from triple-negative breast cancer (TNBC) patients with liver metastasis contain higher TGFβ1 levels and induce adhesion of more breast cancer cells to the 3D human liver-chip than do EVs isolated from healthy donors or nonmetastatic TNBC patients. These findings provide a better understanding of the mechanisms through which breast cancer-derived EVs guide secondary metastasis to the liver. Furthermore, the 3D human liver-chip described in this study provides a platform to investigate the mechanisms underlying secondary metastasis to the liver and possible therapeutic strategies.
Collapse
Affiliation(s)
- Junyoung Kim
- Department of Biomedical Engineering, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Chaeeun Lee
- Department of Biomedical Engineering, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Inun Kim
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Jooyoung Ro
- Department of Biomedical Engineering, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Jungmin Kim
- Department of Biomedical Engineering, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Yoohong Min
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Juhee Park
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Vijaya Sunkara
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Yang-Seok Park
- Department of Biomedical Engineering, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Issac Michael
- Department of Biomedical Engineering, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Young-Ae Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Hee Jin Lee
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Yoon-Kyoung Cho
- Department of Biomedical Engineering, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| |
Collapse
|