1
|
Angeli S, Neophytou C, Kalli M, Stylianopoulos T, Mpekris F. The mechanopathology of the tumor microenvironment: detection techniques, molecular mechanisms and therapeutic opportunities. Front Cell Dev Biol 2025; 13:1564626. [PMID: 40171226 PMCID: PMC11958720 DOI: 10.3389/fcell.2025.1564626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 02/27/2025] [Indexed: 04/03/2025] Open
Abstract
The mechanical properties of the tumor microenvironment (TME) undergo significant changes during tumor growth, primarily driven by alterations in extracellular (ECM) stiffness and tumor viscoelasticity. These mechanical changes not only promote tumor progression but also hinder therapeutic efficacy by impairing drug delivery and activating mechanotransduction pathways that regulate crucial cellular processes such as migration, proliferation, and resistance to therapy. In this review, we examine the mechanisms through which tumor cells sense and transmit mechanical signals to maintain homeostasis in the biomechanically altered TME. We explore current computational modelling strategies for mechanotransduction pathways, highlighting the need for developing models that incorporate additional components of the mechanosignaling machinery. Furthermore, we review available methods for measuring the mechanical properties of tumors in clinical settings and strategies aiming at restoring the TME and blocking deregulated mechanotransduction pathways. Finally, we propose that proper characterization and a deeper understanding of the mechanical landscape of the TME, both at the tissue and cellular levels, are essential for developing therapeutic strategies that account for the influence of mechanical forces on treatment efficacy.
Collapse
Affiliation(s)
| | | | | | | | - Fotios Mpekris
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
2
|
Zhang Y, Rao Y, Lu J, Wang J, Ker DFE, Zhou J, Wang DM. The influence of biophysical niche on tumor-associated macrophages in liver cancer. Hepatol Commun 2024; 8:e0569. [PMID: 39470328 PMCID: PMC11524744 DOI: 10.1097/hc9.0000000000000569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 08/30/2024] [Indexed: 10/30/2024] Open
Abstract
HCC, the most common type of primary liver cancer, is a leading cause of cancer-related mortality worldwide. Although the advancement of immunotherapies by immune checkpoint inhibitors (ICIs) that target programmed cell death 1 or programmed cell death 1-ligand 1 has revolutionized the treatment for HCC, the majority is still not beneficial. Accumulating evidence has pointed out that the potent immunosuppressive tumor microenvironment in HCC poses a great challenge to ICI therapeutic efficacy. As a key component in tumor microenvironment, tumor-associated macrophages (TAMs) play vital roles in HCC development, progression, and ICI low responsiveness. Mechanistically, TAM can promote cancer invasion and metastasis, angiogenesis, epithelial-mesenchymal transition, maintenance of stemness, and most importantly, immunosuppression. Targeting TAMs, therefore, represents an opportunity to enhance the ICI therapeutic efficacy in patients with HCC. While previous research has primarily focused on biochemical cues influencing macrophages, emerging evidence highlights the critical role of biophysical signals, such as substrate stiffness, topography, and external forces. In this review, we summarize the influence of biophysical characteristics within the tumor microenvironment that regulate the phenotype and function of TAMs in HCC pathogenesis and progression. We also explore the possible mechanisms and discuss the potential of manipulating biophysical cues in regulating TAM for HCC therapy. By gaining a deeper understanding of how macrophages sense and respond to mechanical forces, we may potentially usher in a path toward a curative approach for combinatory cancer immunotherapies.
Collapse
Affiliation(s)
- Ying Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Ying Rao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Jiahuan Lu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Jiyu Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Dai Fei Elmer Ker
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Sha Tin, Hong Kong, SAR, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, SAR, China
| | - Jingying Zhou
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Dan Michelle Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Institute of Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Sha Tin, Hong Kong, SAR, China
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| |
Collapse
|
3
|
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
|
4
|
Kalli M, Stylianopoulos T. Toward innovative approaches for exploring the mechanically regulated tumor-immune microenvironment. APL Bioeng 2024; 8:011501. [PMID: 38390314 PMCID: PMC10883717 DOI: 10.1063/5.0183302] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 01/30/2024] [Indexed: 02/24/2024] Open
Abstract
Within the complex tumor microenvironment, cells experience mechanical cues-such as extracellular matrix stiffening and elevation of solid stress, interstitial fluid pressure, and fluid shear stress-that significantly impact cancer cell behavior and immune responses. Recognizing the significance of these mechanical cues not only sheds light on cancer progression but also holds promise for identifying potential biomarkers that would predict therapeutic outcomes. However, standardizing methods for studying how mechanical cues affect tumor progression is challenging. This challenge stems from the limitations of traditional in vitro cell culture systems, which fail to encompass the critical contextual cues present in vivo. To address this, 3D tumor spheroids have been established as a preferred model, more closely mimicking cancer progression, but they usually lack reproduction of the mechanical microenvironment encountered in actual solid tumors. Here, we review the role of mechanical forces in modulating tumor- and immune-cell responses and discuss how grasping the importance of these mechanical cues could revolutionize in vitro tumor tissue engineering. The creation of more physiologically relevant environments that better replicate in vivo conditions will eventually increase the efficacy of currently available treatments, including immunotherapies.
Collapse
Affiliation(s)
- Maria Kalli
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
5
|
Xin Y, Li K, Huang M, Liang C, Siemann D, Wu L, Tan Y, Tang X. Biophysics in tumor growth and progression: from single mechano-sensitive molecules to mechanomedicine. Oncogene 2023; 42:3457-3490. [PMID: 37864030 PMCID: PMC10656290 DOI: 10.1038/s41388-023-02844-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/08/2023] [Accepted: 09/15/2023] [Indexed: 10/22/2023]
Abstract
Evidence from physical sciences in oncology increasingly suggests that the interplay between the biophysical tumor microenvironment and genetic regulation has significant impact on tumor progression. Especially, tumor cells and the associated stromal cells not only alter their own cytoskeleton and physical properties but also remodel the microenvironment with anomalous physical properties. Together, these altered mechano-omics of tumor tissues and their constituents fundamentally shift the mechanotransduction paradigms in tumorous and stromal cells and activate oncogenic signaling within the neoplastic niche to facilitate tumor progression. However, current findings on tumor biophysics are limited, scattered, and often contradictory in multiple contexts. Systematic understanding of how biophysical cues influence tumor pathophysiology is still lacking. This review discusses recent different schools of findings in tumor biophysics that have arisen from multi-scale mechanobiology and the cutting-edge technologies. These findings range from the molecular and cellular to the whole tissue level and feature functional crosstalk between mechanotransduction and oncogenic signaling. We highlight the potential of these anomalous physical alterations as new therapeutic targets for cancer mechanomedicine. This framework reconciles opposing opinions in the field, proposes new directions for future cancer research, and conceptualizes novel mechanomedicine landscape to overcome the inherent shortcomings of conventional cancer diagnosis and therapies.
Collapse
Grants
- R35 GM150812 NIGMS NIH HHS
- This work was financially supported by National Natural Science Foundation of China (Project no. 11972316, Y.T.), Shenzhen Science and Technology Innovation Commission (Project no. JCYJ20200109142001798, SGDX2020110309520303, and JCYJ20220531091002006, Y.T.), General Research Fund of Hong Kong Research Grant Council (PolyU 15214320, Y. T.), Health and Medical Research Fund (HMRF18191421, Y.T.), Hong Kong Polytechnic University (1-CD75, 1-ZE2M, and 1-ZVY1, Y.T.), the Cancer Pilot Research Award from UF Health Cancer Center (X. T.), the National Institute of General Medical Sciences of the National Institutes of Health under award number R35GM150812 (X. T.), the National Science Foundation under grant number 2308574 (X. T.), the Air Force Office of Scientific Research under award number FA9550-23-1-0393 (X. T.), the University Scholar Program (X. T.), UF Research Opportunity Seed Fund (X. T.), the Gatorade Award (X. T.), and the National Science Foundation REU Site at UF: Engineering for Healthcare (Douglas Spearot and Malisa Sarntinoranont). We are deeply grateful for the insightful discussions with and generous support from all members of Tang (UF)’s and Tan (PolyU)’s laboratories and all staff members of the MAE/BME/ECE/Health Cancer Center at UF and BME at PolyU.
- National Natural Science Foundation of China (National Science Foundation of China)
- Shenzhen Science and Technology Innovation Commission
Collapse
Affiliation(s)
- Ying Xin
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Keming Li
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Miao Huang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Chenyu Liang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Dietmar Siemann
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Lizi Wu
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Youhua Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China.
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China.
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Xin Tang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA.
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA.
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
6
|
Juste-Lanas Y, Hervas-Raluy S, García-Aznar JM, González-Loyola A. Fluid flow to mimic organ function in 3D in vitro models. APL Bioeng 2023; 7:031501. [PMID: 37547671 PMCID: PMC10404142 DOI: 10.1063/5.0146000] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/20/2023] [Indexed: 08/08/2023] Open
Abstract
Many different strategies can be found in the literature to model organ physiology, tissue functionality, and disease in vitro; however, most of these models lack the physiological fluid dynamics present in vivo. Here, we highlight the importance of fluid flow for tissue homeostasis, specifically in vessels, other lumen structures, and interstitium, to point out the need of perfusion in current 3D in vitro models. Importantly, the advantages and limitations of the different current experimental fluid-flow setups are discussed. Finally, we shed light on current challenges and future focus of fluid flow models applied to the newest bioengineering state-of-the-art platforms, such as organoids and organ-on-a-chip, as the most sophisticated and physiological preclinical platforms.
Collapse
Affiliation(s)
| | - Silvia Hervas-Raluy
- Department of Mechanical Engineering, Engineering Research Institute of Aragón (I3A), University of Zaragoza, Zaragoza, Spain
| | | | | |
Collapse
|
7
|
Jasuja H, Jaswandkar SV, Katti DR, Katti KS. Interstitial fluid flow contributes to prostate cancer invasion and migration to bone; study conducted using a novel horizontal flow bioreactor. Biofabrication 2023; 15:025017. [PMID: 36863017 PMCID: PMC10020972 DOI: 10.1088/1758-5090/acc09a] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 03/01/2023] [Indexed: 03/04/2023]
Abstract
Prostate cancer bone metastasis is the leading cause of cancer-related mortality in men in the United States, causing severe damage to skeletal tissue. The treatment of advanced-stage prostate cancer is always challenging due to limited drug treatment options, resulting in low survival rates. There is a scarcity of knowledge regarding the mechanisms associated with the effects of biomechanical cues by the interstitial fluid flow on prostate cancer cell growth and migration. We have designed a novel bioreactor system to demonstrate the impact of interstitial fluid flow on the migration of prostate cancer cells to the bone during extravasation. First, we demonstrated that a high flow rate induces apoptosis in PC3 cells via TGF-β1 mediated signaling; thus, physiological flow rate conditions are optimum for cell growth. Next, to understand the role of interstitial fluid flow in prostate cancer migration, we evaluated the migration rate of cells under static and dynamic conditions in the presence or absence of bone. We report that CXCR4 levels were not significantly changed under static and dynamic conditions, indicating that CXCR4 activation in PC3 cells is not influenced by flow conditions but by the bone, where CXCR4 levels were upregulated. The bone-upregulated CXCR4 levels led to increased MMP-9 levels resulting in a high migration rate in the presence of bone. In addition, upregulated levels ofαvβ3integrins under fluid flow conditions contributed to an overall increase in the migration rate of PC3 cells. Overall, this study demonstrates the potential role of interstitial fluid flow in prostate cancer invasion. Understanding the critical role of interstitial fluid flow in promoting prostate cancer cell progression will enhance current therapies for advanced-stage prostate cancer and provide improved treatment options for patients.
Collapse
Affiliation(s)
- Haneesh Jasuja
- Department of Civil, Construction and Environmental Engineering North Dakota State University, Fargo, ND 58108, United States of America
| | - Sharad V Jaswandkar
- Department of Civil, Construction and Environmental Engineering North Dakota State University, Fargo, ND 58108, United States of America
| | - Dinesh R Katti
- Department of Civil, Construction and Environmental Engineering North Dakota State University, Fargo, ND 58108, United States of America
| | - Kalpana S Katti
- Department of Civil, Construction and Environmental Engineering North Dakota State University, Fargo, ND 58108, United States of America
- Author to whom any correspondence should be addressed
| |
Collapse
|
8
|
Akerkouch L, Jasuja H, Katti K, Katti D, Le T. The Influence of Fluid Shear Stress on Bone and Cancer Cells Proliferation and Distribution. Ann Biomed Eng 2023; 51:1199-1215. [PMID: 36593306 DOI: 10.1007/s10439-022-03123-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 12/25/2022] [Indexed: 01/04/2023]
Abstract
We investigated the potential correlation between the fluid shear stress and the proliferation of bone prostate cancer cells on the surface of nanoclay-based scaffolds in a perfusion bioreactor. Human mesenchymal stem cells (hMSCs) were seeded on the scaffolds to initiate bone growth. After 23 days, prostate cancer cells (MDAPCa2b) were cultured on top of the osteogenically differentiated hMSCs. The scaffolds were separated into two groups subjected to two distinct conditions: (i) static (no flow); and (ii) dynamic (with flow) conditions to recapitulate bone metastasis of prostate cancer. Based on measured data, Computational Fluid Dynamics (CFD) models were constructed to determine the velocity and shear stress distributions on the scaffold surface. Our experimental results show distinct differences in the growth pattern of hMSCs and MDAPCa2b cells between the static and dynamic conditions. Our computational results further suggest that the dynamic flow leads to drastic change in cell morphology and tumorous distribution. Our work points to a strong correlation between tumor growth and local interstitial flows in bones.
Collapse
Affiliation(s)
- Lahcen Akerkouch
- Department of Civil, Construction and Environmental Engineering, North Dakota State University, Fargo, ND, USA
| | - Haneesh Jasuja
- Department of Civil, Construction and Environmental Engineering, North Dakota State University, Fargo, ND, USA
| | - Kalpana Katti
- Department of Civil, Construction and Environmental Engineering, North Dakota State University, Fargo, ND, USA
| | - Dinesh Katti
- Department of Civil, Construction and Environmental Engineering, North Dakota State University, Fargo, ND, USA
| | - Trung Le
- Department of Civil, Construction and Environmental Engineering, North Dakota State University, Fargo, ND, USA.
| |
Collapse
|
9
|
Jin P, Munson JM. Fluids and flows in brain cancer and neurological disorders. WIREs Mech Dis 2023; 15:e1582. [PMID: 36000149 PMCID: PMC9869390 DOI: 10.1002/wsbm.1582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 06/21/2022] [Accepted: 06/27/2022] [Indexed: 01/31/2023]
Abstract
Interstitial fluid (IF) and cerebrospinal fluid (CSF) are an integral part of the brain, serving to cushion and protect the brain parenchymal cells against damage and aid in their function. The brain IF contains various ions, nutrients, waste products, peptides, hormones, and neurotransmitters. IF moves primarily by pressure-dependent bulk flow through brain parenchyma, draining into the ventricular CSF. The brain ventricles and subarachnoid spaces are filled with CSF which circulates through the perivascular spaces. It also flows into the IF space regulated, in part, by aquaporin channels, removing waste solutes through a process of IF-CSF mixing. During disease development, the composition, flow, and volume of these fluids changes and can lead to brain cell dysfunction. With the improvement of imaging technology and the help of genomic profiling, more information has been and can be obtained from brain fluids; however, the role of CSF and IF in brain cancer and neurobiological disease is still limited. Here we outline recent advances of our knowledge of brain fluid flow in cancer and neurodegenerative disease based on our understanding of its dynamics and composition. This article is categorized under: Cancer > Biomedical Engineering Neurological Diseases > Biomedical Engineering.
Collapse
Affiliation(s)
- Peng Jin
- Fralin Biomedical Research Institute, Department of Biomedical Engineering and Mechanics Virginia Polytechnic Institute and State University Roanoke Virginia USA
| | - Jennifer M. Munson
- Fralin Biomedical Research Institute, Department of Biomedical Engineering and Mechanics Virginia Polytechnic Institute and State University Roanoke Virginia USA
| |
Collapse
|
10
|
Simchick G, Hernando D. Precision of region of interest-based tri-exponential intravoxel incoherent motion quantification and the role of the Intervoxel spatial distribution of flow velocities. Magn Reson Med 2022; 88:2662-2678. [PMID: 35968580 PMCID: PMC9529845 DOI: 10.1002/mrm.29406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/27/2022] [Accepted: 07/18/2022] [Indexed: 11/08/2022]
Abstract
PURPOSE The purpose of this work was to obtain precise tri-exponential intravoxel incoherent motion (IVIM) quantification in the liver using 2D (b-value and first-order motion moment [M1 ]) IVIM-DWI acquisitions and region of interest (ROI)-based fitting techniques. METHODS Diffusion MRI of the liver was performed in 10 healthy volunteers using three IVIM-DWI acquisitions: conventional monopolar, optimized monopolar, and optimized 2D (b-M1 ). For each acquisition, bi-exponential and tri-exponential full, segmented, and over-segmented ROI-based fitting and a newly proposed blood velocity SDdistribution (BVD) fitting technique were performed to obtain IVIM estimates in the right and left liver lobes. Fitting quality was evaluated using corrected Akaike information criterion. Precision metrics (test-retest repeatability, inter-reader reproducibility, and inter-lobar agreement) were evaluated using Bland-Altman analysis, repeatability/reproducibility coefficients (RPCs), and paired sample t-tests. Precision was compared across acquisitions and fitting methods. RESULTS High repeatability and reproducibility was observed in the estimations of the diffusion coefficient (Dtri = [1.03 ± 0.11] × 10-3 mm2 /s; RPCs ≤ 1.34 × 10-4 mm2 /s), perfusion fractions (F1 = 3.19 ± 1.89% and F2 = 16.4 ± 2.07%; RPCs ≤ 2.51%), and blood velocity SDs (Vb,1 = 1.44 ± 0.14 mm/s and Vb,2 = 3.62 ± 0.13 mm/s; RPCs ≤ 0.41 mm/s) in the right liver lobe using the 2D (b-M1 ) acquisition in conjunction with BVD fitting. Using these methods, significantly larger (p < 0.01) estimates of Dtri and F1 were observed in the left lobe in comparison to the right lobe, while estimates of Vb,1 and Vb,2 demonstrated high interlobar agreement (RPCs ≤ 0.45 mm/s). CONCLUSIONS The 2D (b-M1 ) IVIM-DWI data acquisition in conjunction with BVD fitting enables highly precise tri-exponential IVIM quantification in the right liver lobe.
Collapse
Affiliation(s)
- Gregory Simchick
- Radiology, University of Wisconsin-Madison, Madison, WI, United States
- Medical Physics, University of Wisconsin-Madison, Madison, WI, United States
| | - Diego Hernando
- Radiology, University of Wisconsin-Madison, Madison, WI, United States
- Medical Physics, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
11
|
Chemokine/GPCR Signaling-Mediated EMT in Cancer Metastasis. JOURNAL OF ONCOLOGY 2022; 2022:2208176. [PMID: 36268282 PMCID: PMC9578795 DOI: 10.1155/2022/2208176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 08/08/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022]
Abstract
Metastasis, the chief cause of cancer-related deaths, is associated with epithelial-mesenchymal transition (EMT). In the tumor microenvironment, EMT can be triggered by chemokine/G-protein-coupled receptor (GPCR) signaling, which is closely associated with tumor progression. However, the functional links between chemokine/GPCR signaling-mediated EMT and metastasis remain unclear. Herein, we summarized the mechanisms of chemokine/GPCR signaling-mediated EMT with an insight into facilitating metastasis and clarified the role of chemokine in the local invasion, intravasation, circulation, extravasation, and colonization, respectively. Moreover, several potential pathways that might contribute to EMT based on the latest studies on GPCR signaling were proposed, including signaling mediated by G protein, β-arrestin, intracellular, dimerization activation, and transactivation. However, there is still limited evidence to support the EMT programme functional contribution to metastasis, which keeps a key question still open whether we should target EMT programme of cancer cells. Answers to that question might help develop an anticancer strategy or guide new directions for anticancer metastasis therapy.
Collapse
|
12
|
Li N, Zhang X, Zhou J, Li W, Shu X, Wu Y, Long M. Multiscale biomechanics and mechanotransduction from liver fibrosis to cancer. Adv Drug Deliv Rev 2022; 188:114448. [PMID: 35820602 DOI: 10.1016/j.addr.2022.114448] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/08/2022] [Accepted: 07/06/2022] [Indexed: 02/06/2023]
Abstract
A growing body of multiscale biomechanical studies has been proposed to highlight the mechanical cues in the development of hepatic fibrosis and cancer. At the cellular level, changes in mechanical microenvironment induce phenotypic and functional alterations of hepatic cells, initiating a positive feedback loop that promotes liver fibrogenesis and hepatocarcinogenesis. Tumor mechanical microenvironment of hepatocellular carcinoma facilitates tumor cell growth and metastasis, and hinders the drug delivery and immunotherapy. At the molecular level, mechanical forces are sensed and transmitted into hepatic cells via allosteric activation of mechanoreceptors on the cell membrane, leading to the activation of various mechanotransduction pathways including integrin and YAP signaling and then regulating cell function. Thus, the application of mechanomedicine concept in the treatment of liver diseases is promising for rational design and cell-specific delivery of therapeutic drugs. This review mainly discusses the correlation between biomechanical cues and liver diseases from the viewpoint of mechanobiology.
Collapse
Affiliation(s)
- Ning Li
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoyu Zhang
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jin Zhou
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
| | - Wang Li
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinyu Shu
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi Wu
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mian Long
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
13
|
Roberts LM, Perez MJ, Balogh KN, Mingledorff G, Cross JV, Munson JM. Myeloid Derived Suppressor Cells Migrate in Response to Flow and Lymphatic Endothelial Cell Interaction in the Breast Tumor Microenvironment. Cancers (Basel) 2022; 14:cancers14123008. [PMID: 35740673 PMCID: PMC9221529 DOI: 10.3390/cancers14123008] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/26/2022] [Accepted: 06/07/2022] [Indexed: 12/07/2022] Open
Abstract
At the site of the tumor, myeloid derived suppressor cells (MDSCs) infiltrate and interact with elements of the tumor microenvironment in complex ways. Within the invading tumor, MDSCs are exposed to interstitial fluid flow (IFF) that exists within the chronic inflammatory tumor microenvironment at the tumor-lymphatic interface. As drivers of cell migration and invasion, the link between interstitial fluid flow, lymphatics, and MDSCs have not been clearly established. Here, we hypothesized that interstitial fluid flow and cells within the breast tumor microenvironment modulate migration of MDSCs. We developed a novel 3D model to mimic the breast tumor microenvironment and incorporated MDSCs harvested from 4T1-tumor bearing mice. Using live imaging, we found that sorted GR1+ splenocytes had reduced chemotactic index compared to the unsorted population, but their speed and displacement were similar. Using our adapted tissue culture insert assay, we show that interstitial fluid flow promotes MDSC invasion, regardless of absence or presence of tumor cells. Coordinating with lymphatic endothelial cells, interstitial fluid flow further enhanced invasion of MDSCs in the presence of 4T1 cells. We also show that VEGFR3 inhibition reduced both MDSC and 4T1 flow response. Together, these findings indicate a key role of interstitial fluid flow in MDSC migration as well as describe a tool to explore the immune microenvironment in breast cancer.
Collapse
Affiliation(s)
- LaDeidra Monét Roberts
- Department of Biomedical Engineering and Mechanics, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA 24016, USA;
| | - Matthew J. Perez
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22904, USA;
| | - Kristen N. Balogh
- Department of Pathology, University of Virginia, Charlottesville, VA 22904, USA; (K.N.B.); (J.V.C.)
| | - Garnett Mingledorff
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22904, USA;
| | - Janet V. Cross
- Department of Pathology, University of Virginia, Charlottesville, VA 22904, USA; (K.N.B.); (J.V.C.)
| | - Jennifer M. Munson
- Department of Biomedical Engineering and Mechanics, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA 24016, USA;
- Correspondence:
| |
Collapse
|
14
|
Song M, Kuerban M, Zhao L, Peng X, Xu Y. Inhibition of RFX6 Suppresses the Invasive Ability of Tumor Cells Through the Notch Pathway and Affects Tumor Immunity in Hepatocellular Carcinoma. Front Oncol 2022; 11:801222. [PMID: 34988028 PMCID: PMC8721116 DOI: 10.3389/fonc.2021.801222] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/23/2021] [Indexed: 11/23/2022] Open
Abstract
Background The DNA-binding protein RFX6 was overexpressed in hepatocellular carcinoma, and its expression level was correlated with the prognosis and immune cell infiltration in liver hepatocellular carcinoma. However, the mechanism of the abnormal expression and the biological effects of RFX6 in liver cancer remains unknown. Methods To understand the specific expression mechanism of RFX6 in liver cancer, we performed bioinformatic prediction, CHIP-qPCR assay, co-IP, and dual-luciferase assay to assess the regulating mechanism of RFX6. In the meantime, a series of biological experiments in vivo and in vitro were conducted to analyze the biological significance of RFX6 in hepatocellular carcinoma. Results We demonstrated that knockdown of RFX6 in liver cancer cells significantly suppressed the proliferation, migration, and invasion of cancer cells. Moreover, inhibition of RFX6 could affect the immune response of T cells. Among a number of interacting proteins, we revealed that RFX6 directly binds to DTX2, a regulator of the Notch signaling pathway by targeting NOTCH1, and helps in its transcription stability. Furthermore, we discovered that miRNA-542-3p, the expression of which was decreased in hepatocellular carcinoma, was directly involved in the negative regulation of the expression of RFX6. Conclusion In summary, we discovered that the miRNA-542-3p–RFX6–DTX2–NOTCH1 regulatory pathway played significant roles in the tumor progression of liver hepatocellular carcinoma.
Collapse
Affiliation(s)
- Mu Song
- Department of Surgical Oncology, The Second Affiliated Hospital, Xinjiang Medical University, Urumqi, China.,Department of Thyroid and Breast Surgery, The Seventh Affiliated Hospital, Southern Medical University, Foshan, China
| | - Mulati Kuerban
- Department of Surgical Oncology, The Seventh Affiliated Hospital, Xinjiang Medical University, Urumqi, China
| | - Lu Zhao
- Department of Surgical Oncology, The Second Affiliated Hospital, Xinjiang Medical University, Urumqi, China.,Department of Thyroid and Breast Surgery, The Seventh Affiliated Hospital, Southern Medical University, Foshan, China
| | - Xiaolin Peng
- Department of Thyroid and Breast Surgery, The Seventh Affiliated Hospital, Southern Medical University, Foshan, China
| | - Youqin Xu
- Department of Thyroid and Breast Surgery, The Seventh Affiliated Hospital, Southern Medical University, Foshan, China
| |
Collapse
|
15
|
Tian BR, Lin WF, Zhang Y. Effects of biomechanical forces on the biological behavior of cancer stem cells. J Cancer 2021; 12:5895-5902. [PMID: 34476003 PMCID: PMC8408108 DOI: 10.7150/jca.60893] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 07/20/2021] [Indexed: 12/13/2022] Open
Abstract
Cancer stem cells (CSCs), dynamic subsets of cancer cells, are responsible for malignant progression. The unique properties of CSCs, including self-renewal, differentiation, and malignancy, closely depend on the tumor microenvironment. Mechanical components in the microenvironment, including matrix stiffness, fluid shear stress, compression and tension stress, affect the fate of CSCs and further influence the cancer process. This paper reviews recent studies of mechanical components and CSCs, and further discusses the intrinsic correlation among them. Regulatory mechanisms of mechanical microenvironment, which act on CSCs, have great potential for clinical application and provide different perspectives to drugs and treatment design.
Collapse
Affiliation(s)
- Bo Ren Tian
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, Guangdong, People's Republic of China
| | - Wei Fan Lin
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, Guangdong, People's Republic of China
| | - Yan Zhang
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, Guangdong, People's Republic of China
| |
Collapse
|
16
|
Wang Z, Ao X, Shen Z, Ao L, Wu X, Pu C, Guo W, Xing W, He M, Yuan H, Yu J, Li L, Xu X. TNF-α augments CXCL10/CXCR3 axis activity to induce Epithelial-Mesenchymal Transition in colon cancer cell. Int J Biol Sci 2021; 17:2683-2702. [PMID: 34345201 PMCID: PMC8326125 DOI: 10.7150/ijbs.61350] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/13/2021] [Indexed: 12/18/2022] Open
Abstract
Chronic inflammation-induced metastases have long been regarded as one of the significant obstacles in treating cancer. Tumor necrosis factor-α (TNF-α), a main inflammation mediator within tumor microenvironment, affects tumor development by inducing multiple chemokines to establish a complex network. Recent reports have revealed that CXCL10/CXCR3 axis affects cancer cells invasiveness and metastases, and Epithelial-mesenchymal transition (EMT) is the main reason for frequent proliferation and distant organ metastases of colon cancer (CC) cells, However, it is unclear whether TNF-α- mediated chronic inflammation can synergically enhance EMT-mediated CC metastasis through promoting chemokine expression. According to this study, TNF-α activated the PI3K/Akt and p38 MAPK parallel signal transduction pathways, then stimulate downstream NF-κB pathway p65 into the nucleus to activate CXCL10 transcription. CXCL10 enhanced the metastases of CC-cells by triggering small GTPases such as RhoA and cdc42. Furthermore, overexpression of CXCL10 significantly enhanced tumorigenicity and mobility of CC cells in vivo. We further clarified that CXCL10 activated the PI3K/Akt pathway through CXCR3, resulting in suppression of GSK-3β phosphorylation and leading to upregulation of Snail expression, thereby regulating EMT in CC cells. These outcomes lay the foundation for finding new targets to inhibit CC metastases.
Collapse
Affiliation(s)
- Zhengcheng Wang
- Department of Human Anatomy and Histology and Embryology, School of Basic Medical Sciences, Qingdao University, Qingdao 266000, China
| | - Xiang Ao
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Zhilin Shen
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Luoquan Ao
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Xiaofeng Wu
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Chengxiu Pu
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Wei Guo
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Wei Xing
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Min He
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Hongfeng Yuan
- Department of Ophthalmology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Jianhua Yu
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Ling Li
- Department of Human Anatomy and Histology and Embryology, School of Basic Medical Sciences, Qingdao University, Qingdao 266000, China
| | - Xiang Xu
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| |
Collapse
|
17
|
CXCL2/10/12/14 are prognostic biomarkers and correlated with immune infiltration in hepatocellular carcinoma. Biosci Rep 2021; 41:228875. [PMID: 34085699 PMCID: PMC8217985 DOI: 10.1042/bsr20204312] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 05/16/2021] [Accepted: 06/02/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND C-x-C motif chemokine ligands (CXCLs) are critical regulators of cancer immunity and angiogenesis, which affect disease progression and treatment responses. The character of each CXCL in the prognosis and immune infiltration of hepatocellular carcinoma (HCC) patients is unclear yet. METHODS Differentially expressed CXCLs between HCC and normal control were screened by Oncomine and GEPIA2. Genetic alternations of CXCLs in HCC were analyzed by cBioPortal. Clinicopathological relevance of CXCLs in HCC patients was analyzed using UALCAN. The prognostic value of CXCLs was evaluated using univariate and multivariate analyses. Correlations of CXCLs' expression with immune infiltration, chemokines and their receptors were assessed integrating TIMER, TISIDB, and GEPIA2. The co-expressed genes of CXCLs were discovered, and functional enrichment analysis was performed for them. RESULTS CXCL9/10 was significantly higher expressed while CXCL2/12/14 was lower expressed in HCC than normal tissues, but they didn't show significant clinicopathological relevance in HCC patients. High-expression of CXCL2/10/12/14 indicated favorable outcomes of HCC patients. The expression of CXCL9/10/12/14 was significantly positively correlated with not only the infiltration and biomarkers' expression of various tumor-infiltrating immune cells but also the abundance of chemokines and their receptors. The co-expressed genes of the five CXCLs were extracellular components and regulated immune or inflammatory responses and signaling pathways of chemokine, Toll-like receptor and tumor necrosis factor might be involved. CONCLUSION The present study proposed CXCL2/10/12/14 might predict outcomes of HCC patients and were extensively related with the immune microenvironment in HCC. It would be a prospective therapeutic strategy for HCC to enhance effective immunity surveillance through intervening in these CXCLs.
Collapse
|
18
|
Pagé G, Tardieu M, Gennisson JL, Besret L, Garteiser P, Van Beers BE. Tumor Solid Stress: Assessment with MR Elastography under Compression of Patient-Derived Hepatocellular Carcinomas and Cholangiocarcinomas Xenografted in Mice. Cancers (Basel) 2021; 13:cancers13081891. [PMID: 33920771 PMCID: PMC8071192 DOI: 10.3390/cancers13081891] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/09/2021] [Accepted: 04/13/2021] [Indexed: 01/01/2023] Open
Abstract
Malignant tumors have abnormal biomechanical characteristics, including high viscoelasticity, solid stress, and interstitial fluid pressure. Magnetic resonance (MR) elastography is increasingly used to non-invasively assess tissue viscoelasticity. However, solid stress and interstitial fluid pressure measurements are performed with invasive methods. We studied the feasibility and potential role of MR elastography at basal state and under controlled compression in assessing altered biomechanical features of malignant liver tumors. MR elastography was performed in mice with patient-derived, subcutaneously xenografted hepatocellular carcinomas or cholangiocarcinomas to measure the basal viscoelasticity and the compression stiffening rate, which corresponds to the slope of elasticity versus applied compression. MR elastography measurements were correlated with invasive pressure measurements and digital histological readings. Significant differences in MR elastography parameters, pressure, and histological measurements were observed between tumor models. In multivariate analysis, collagen content and interstitial fluid pressure were determinants of basal viscoelasticity, whereas solid stress, in addition to collagen content, cellularity, and tumor type, was an independent determinant of compression stiffening rate. Compression stiffening rate had high AUC (0.87 ± 0.08) for determining elevated solid stress, whereas basal elasticity had high AUC for tumor collagen content (AUC: 0.86 ± 0.08). Our results suggest that MR elastography compression stiffening rate, in contrast to basal viscoelasticity, is a potential marker of solid stress in malignant liver tumors.
Collapse
Affiliation(s)
- Gwenaël Pagé
- Laboratory of Imaging Biomarkers, Center of Research on Inflammation, Université de Paris, UMR 1149, Inserm, F-75018 Paris, France; (P.G.); (B.E.V.B.)
- Correspondence:
| | - Marion Tardieu
- Montpellier Cancer Research Institute (IRCM), INSERM U1194, University of Montpellier, 34095 Montpellier, France;
- Montpellier Cancer Institute (ICM), 34298 Montpellier, France
| | - Jean-Luc Gennisson
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, 91401 Orsay, France;
| | | | - Philippe Garteiser
- Laboratory of Imaging Biomarkers, Center of Research on Inflammation, Université de Paris, UMR 1149, Inserm, F-75018 Paris, France; (P.G.); (B.E.V.B.)
| | - Bernard E. Van Beers
- Laboratory of Imaging Biomarkers, Center of Research on Inflammation, Université de Paris, UMR 1149, Inserm, F-75018 Paris, France; (P.G.); (B.E.V.B.)
- Department of Radiology, AP-HP, Beaujon University Hospital Paris Nord, F-92110 Clichy, France
| |
Collapse
|
19
|
Park SJ, Yoon JH, Lee DH, Lim WH, Lee JM. Tumor Stiffness Measurements on MR Elastography for Single Nodular Hepatocellular Carcinomas Can Predict Tumor Recurrence After Hepatic Resection. J Magn Reson Imaging 2020; 53:587-596. [PMID: 32914909 DOI: 10.1002/jmri.27359] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 08/27/2020] [Accepted: 08/27/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Tumor stiffness (TS), measured by magnetic resonance elastography (MRE), could be associated with tumor mechanical properties and tumor grade. PURPOSE To determine whether TS obtained using MRE is associated with survival in patients with single nodular hepatocellular carcinoma (HCC) after hepatic resection (HR). STUDY TYPE Retrospective. POPULATION In all, 95 patients with pathologically confirmed HCCs. FIELD STRENGTH/SEQUENCE 1.5T/3D spin-echo echo-planar imaging MRE. ASSESSMENT TS values of the whole tumor (TS-WT) and of a solid portion of the tumor (TS-SP) after excluding the necrotic area were measured on stiffness maps. Known imaging prognostic factors of HCC were also analyzed. After surgery, pathologic findings were evaluated from resected pathology specimens. STATISTICAL TESTS Fisher's exact test and the Mann-Whitney U-test were performed to determine the significance of differences according to the tumor grade. Overall survival (OS) / recurrence-free survival (RFS) analyses were performed using Kaplan-Meier analyses and Cox multivariable models. RESULTS The average TS-WT was 2.14 ± 0.74 kPa, and the average TS-SP was 2.51 ± 1.07 kPa. The cumulative incidence of RFS was 73.1%, 63.1%, and 57.3% at 1, 3, and 5 years, respectively. The TS-WT, TS-SP, and tumor size (≥5 cm) were significant prognostic factors for RFS (P < 0.001; P < 0.001; P = 0.017, respectively). The estimated overall 1-, 3-, and 5-year survival rates were 95.7%, 86.9%, and 80.8%, respectively. The alpha-fetoprotein changes, platelets, tumor size (≥5 cm), and vascular invasion in pathology were significant predictive factors for overall survival (all P < 0.05). Tumor necrosis, TS-WT, TS-SP, and vascular invasion in pathology were significantly correlated with poorly differentiated HCC (all P < 0.05). DATA CONCLUSION The TS-WT, TW-SP, and tumor size (≥5 cm) were significant predictive factors of RFS after HR in patients with HCC. Level of Evidence Technical Efficacy Stage 5 J. MAGN. RESON. IMAGING 2021;53:587-596.
Collapse
Affiliation(s)
- Sae-Jin Park
- Department of Radiology, Seoul National University Hospital, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| | - Jeong Hee Yoon
- Department of Radiology, Seoul National University Hospital, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| | - Dong Ho Lee
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
| | - Woo Hyeon Lim
- Department of Radiology, Seoul National University Hospital, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| | - Jeong Min Lee
- Department of Radiology, Seoul National University Hospital, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea.,Institute of Radiation Medicine, Seoul National University Medical Research Center, Seoul, Korea
| |
Collapse
|
20
|
Huang YL, Ma Y, Wu C, Shiau C, Segall JE, Wu M. Tumor spheroids under perfusion within a 3D microfluidic platform reveal critical roles of cell-cell adhesion in tumor invasion. Sci Rep 2020; 10:9648. [PMID: 32541776 PMCID: PMC7295764 DOI: 10.1038/s41598-020-66528-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 05/11/2020] [Indexed: 12/20/2022] Open
Abstract
Tumor invasion within the interstitial space is critically regulated by the force balance between cell-extracellular matrix (ECM) and cell-cell interactions. Interstitial flows (IFs) are present in both healthy and diseased tissues. However, the roles of IFs in modulating cell force balance and subsequently tumor invasion are understudied. In this article, we develop a microfluidic model in which tumor spheroids are embedded within 3D collagen matrices with well-defined IFs. Using co-cultured tumor spheroids (1:1 mixture of metastatic and non-tumorigenic epithelial cells), we show that IFs downregulate the cell-cell adhesion molecule E-cadherin on non-tumorigenic cells and promote tumor invasion. Our microfluidic model advances current tumor invasion assays towards a more physiologically realistic model using tumor spheroids instead of single cells under perfusion. We identify a novel mechanism by which IFs can promote tumor invasion through an influence on cell-cell adhesion within the tumor and highlight the importance of biophysical parameters in regulating tumor invasion.
Collapse
Affiliation(s)
- Yu Ling Huang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Yujie Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Cindy Wu
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Carina Shiau
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Jeffrey E Segall
- Anatomy and Structural Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, 10461, New York, USA
| | - Mingming Wu
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
21
|
SOX4 activates CXCL12 in hepatocellular carcinoma cells to modulate endothelial cell migration and angiogenesis in vivo. Oncogene 2020; 39:4695-4710. [PMID: 32404985 DOI: 10.1038/s41388-020-1319-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 02/07/2023]
Abstract
The overexpression of SOX4 in various kinds of cancer cells was associated with poor prognosis for patients. The role of SOX4 in angiogenesis and tumor microenvironment modulation was recently documented in breast cancer but remains unclear in hepatocellular carcinoma (HCC). In our study, the clinical relevance of SOX4 overexpression in HCC and its role in the tumor microenvironment were investigated. The overexpression of SOX4 (SOX4high) in tumor lesions was associated with higher microvessel density (P = 0.012), tumor thrombosis formation (P = 0.012), distant metastasis (P < 0.001), and an independent prognostic factor for disease-free survival in HCC patients (P = 0.048). Endogenous SOX4 knockout in Hep3B cells by the CRISPR/cas9 system reduced the expression of CXCL12, which, in turn, attenuated chemotaxis in human umbilical vein endothelial cells, tube formation in vitro, reduced tumor growth, reticular fiber production, and angiogenesis in vivo in a xenograft mouse model. Treatment with an antagonist targeting CXCR4 (AMD3100), a receptor of CXCL12, inhibited chemotaxis and tube formation in endothelial cells in vitro. The CXCL12 promoter was activated by ectopic expression of a Flag-tagged SOX4 plasmid, endogenous SOX4 knockdown abolished promoter activity of CXCL12 as shown by luciferase assays, and an association with the CXCL12 promoter was identified via chromatin immunoprecipitation in HCC cells. In conclusion, SOX4 modulates the CXCL12 promoter in HCC cells. The secretory CXCL12, in turn, modulates CXCR4 in endothelial cells, reticular fibers to regulate the tumor microenvironment and modulate neovascularization, which might contribute to the distant metastasis of tumors.
Collapse
|
22
|
Wang H, Mao J, Huang Y, Zhang J, Zhong L, Wu Y, Huang H, Yang J, Wei Y, Tang J. Prognostic roles of miR-124-3p and its target ANXA7 and their effects on cell migration and invasion in hepatocellular carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:357-370. [PMID: 32269673 PMCID: PMC7137028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 02/07/2020] [Indexed: 06/11/2023]
Abstract
Recent studies have indicated that ANXA7 promotes progression and metastasis of hepatocellular carcinoma (HCC). In this study we found a significant negative correlation between the levels of miR-124-3p and ANXA7 protein in HCC. Level of miR-124-3p in tumor tissues was negatively correlated, while ANXA7 protein was positively correlated, with TNM stage and tumor metastasis. Furthermore, we confirmed ANXA7 was a target gene of miR-124-3p by a dual luciferase reporter assay. In vitro, up-regulation of miR-124-3p promotes apoptosis and inhibits migration and invasion of Hca-F. Bcl-2 correlates X protein (Bax) protein level was up-regulated, while ANXA7, B-cell lymphoma-2 (Bcl-2), Matrix metalloproteinase (MMP-9) and C-X-C motif chemokine 12 (CXCL12) protein levels were suppressed relative to miR-124-3p over-expression. In vivo, up-regulation of miR-124-3p suppresses lymph node metastasis (LNM) and tumorigenicity of Hca-F cells. The expression of ANXA7, MMP-9, and CXCL12 protein in transplanted tumors was suppressed relative to miR-124-3p overexpression. In addition, we found the levels of Bcl-2, MMP-9, and CXCL12 in Hca-F cells decreased significantly after transfection of shRNA-Anxa7 in vitro. In conclusion, our study revealed miR-124-3p inhibits tumor growth, invasion, and lymphatic metastasis in HCC by down-regulation of ANXA7 gene, thereby reducing the expression of Bcl-2, MMP-9, and CXCL12.
Collapse
Affiliation(s)
- Honghai Wang
- Department of Pathology, College of Basic Medical Sciences, Dalian Medical UniversityDalian, Liaoning Province, China
| | - Jun Mao
- Teaching Laboratory of Morphology, College of Basic Medical Sciences, Dalian Medical UniversityDalian, Liaoning Province, China
| | - Yuhong Huang
- Department of Pathology, College of Basic Medical Sciences, Dalian Medical UniversityDalian, Liaoning Province, China
- Key Laboratory for Tumor Metastasis and Intervention of Liaoning ProvinceDalian, Liaoning Province, China
| | - Jun Zhang
- Department of Pathology, College of Basic Medical Sciences, Dalian Medical UniversityDalian, Liaoning Province, China
- Key Laboratory for Tumor Metastasis and Intervention of Liaoning ProvinceDalian, Liaoning Province, China
| | - Lin Zhong
- Department of Pathology, The First Affiliated Hospital of Dalian Medical UniversityDalian, China
| | - Ying Wu
- Department of Pathology, College of Basic Medical Sciences, Dalian Medical UniversityDalian, Liaoning Province, China
| | - He Huang
- Department of Pathology, College of Basic Medical Sciences, Dalian Medical UniversityDalian, Liaoning Province, China
| | - Jiayu Yang
- Department of Pathology, College of Basic Medical Sciences, Dalian Medical UniversityDalian, Liaoning Province, China
| | - Yuanyi Wei
- Department of Pathology, College of Basic Medical Sciences, Dalian Medical UniversityDalian, Liaoning Province, China
- Key Laboratory for Tumor Metastasis and Intervention of Liaoning ProvinceDalian, Liaoning Province, China
| | - Jianwu Tang
- Department of Pathology, College of Basic Medical Sciences, Dalian Medical UniversityDalian, Liaoning Province, China
- Key Laboratory for Tumor Metastasis and Intervention of Liaoning ProvinceDalian, Liaoning Province, China
| |
Collapse
|
23
|
Liu L, Jiang H, Zhao W, Meng Y, Li J, Huang T, Sun J. Cdc42-mediated supracellular cytoskeleton induced cancer cell migration under low shear stress. Biochem Biophys Res Commun 2019; 519:134-140. [PMID: 31477271 DOI: 10.1016/j.bbrc.2019.08.149] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 08/26/2019] [Indexed: 01/29/2023]
Abstract
Tumor microenvironment is composed of biological, chemical and physical factors. Mechanical factors are more and more focused these years. Therefore, mimicking mechanical factors' contribution to cancer cell malignancy will greatly improve the advance in this field. Although the induced malignant behaviors are present under many stimuli such as growth or inflammatory factors, the cell key physical migration mechanisms are still missing. In this study, we identify that low shear stress significantly promotes the formation of needle-shaped membrane protrusions, which is called filopodia and important for the sense and interact of a cell with extracellular matrix in the tumor microenvironment. Under low shear stress, the migration is promoted while it is inhibited in the presence of ROCK inhibitor Y27632, which could abolish the F-actin network. Using cell imaging, we further unravel that key to these protrusions is Cell division cycle 42 (Cdc42) dependent. After Cdc42 activation, the filopodia is more and longer, acting as massagers to pass the information from a cell to the microenvironment for its malignant phenotype. In the Cdc42 inhibition, the filopodia is greatly reduced. Moreover, small GTPases Cdc42 rather than Rac1 and Rho directly controls the filopodia formation. Our work highlights that low shear stress and Cdc42 activation are sufficient to promote filopodia formation, it not only points out the novel structure for cancer progression but also provides the experimental physical basis for the efficient drug anti-cancer strategies.
Collapse
Affiliation(s)
- Lingling Liu
- School of Laboratory Medicine, Collaborative Innovation Center of Sichuan for Elderly Care and Health, Development and Regeneration Key Laboratory of Sichuan Province, Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Hua Jiang
- Department of Pediatrics, Department of Microbiology and Immunology, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Wei Zhao
- School of Laboratory Medicine, Collaborative Innovation Center of Sichuan for Elderly Care and Health, Development and Regeneration Key Laboratory of Sichuan Province, Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Yao Meng
- School of Laboratory Medicine, Collaborative Innovation Center of Sichuan for Elderly Care and Health, Development and Regeneration Key Laboratory of Sichuan Province, Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Ji Li
- Department of Clinical Laboratory, Sichuan GEM Flower Hospital, Chengdu, 610213, Sichuan, PR China
| | - Tongwei Huang
- Department of Blood Transfusion, Ziyang Hospital of Traditional Chinese Medicine, Ziyang, 641300, Sichuan, PR China
| | - Jinghui Sun
- School of Laboratory Medicine, Collaborative Innovation Center of Sichuan for Elderly Care and Health, Development and Regeneration Key Laboratory of Sichuan Province, Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu Medical College, Chengdu, 610500, Sichuan, China.
| |
Collapse
|
24
|
Chen W, Zhang J, Fan HN, Zhu JS. Function and therapeutic advances of chemokine and its receptor in nonalcoholic fatty liver disease. Therap Adv Gastroenterol 2018; 11:1756284818815184. [PMID: 30574191 PMCID: PMC6295708 DOI: 10.1177/1756284818815184] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 10/24/2018] [Indexed: 02/04/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) represents a spectrum of hepatic pathology, ranging from simple accumulation of fat in its most benign form, steatohepatitis, to cirrhosis in its most advanced form. The prevalence of NAFLD is 20-30% in adults, and 10-20% of patients with NAFLD progress to nonalcoholic steatohepatitis (NASH) which is predicted to be the leading cause of liver transplantation over the next 10 years. Therefore, it is essential to explore effective diagnostic and treatment strategies for NAFLD patients. Chemokines are a family of small and highly conserved proteins (molecular weight ranging from 8 to 12 kDa) involved in regulating the migration and activities of hepatocytes, Kupffer cells (KCs), hepatic stellate cells (HSCs), endothelial cells and circulating immune cells. Accumulating data show that chemokine and its receptor act vital roles in the pathogenesis of NAFLD. Herein, we summarize the involvement of the chemokine and its receptor in the pathogenesis of NAFLD and explore the novel pharmacotherapeutic avenues for patients with NAFLD.
Collapse
Affiliation(s)
- Wei Chen
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Jing Zhang
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Hui-Ning Fan
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | | |
Collapse
|
25
|
Yeldag G, Rice A, Del Río Hernández A. Chemoresistance and the Self-Maintaining Tumor Microenvironment. Cancers (Basel) 2018; 10:E471. [PMID: 30487436 PMCID: PMC6315745 DOI: 10.3390/cancers10120471] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 11/24/2018] [Accepted: 11/27/2018] [Indexed: 12/15/2022] Open
Abstract
The progression of cancer is associated with alterations in the tumor microenvironment, including changes in extracellular matrix (ECM) composition, matrix rigidity, hypervascularization, hypoxia, and paracrine factors. One key malignant phenotype of cancer cells is their ability to resist chemotherapeutics, and elements of the ECM can promote chemoresistance in cancer cells through a variety of signaling pathways, inducing changes in gene expression and protein activity that allow resistance. Furthermore, the ECM is maintained as an environment that facilitates chemoresistance, since its constitution modulates the phenotype of cancer-associated cells, which themselves affect the microenvironment. In this review, we discuss how the properties of the tumor microenvironment promote chemoresistance in cancer cells, and the interplay between these external stimuli. We focus on both the response of cancer cells to the external environment, as well as the maintenance of the external environment, and how a chemoresistant phenotype emerges from the complex signaling network present.
Collapse
Affiliation(s)
- Gulcen Yeldag
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, UK.
| | - Alistair Rice
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, UK.
| | - Armando Del Río Hernández
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, UK.
| |
Collapse
|
26
|
Kingsmore KM, Vaccari A, Abler D, Cui SX, Epstein FH, Rockne RC, Acton ST, Munson JM. MRI analysis to map interstitial flow in the brain tumor microenvironment. APL Bioeng 2018; 2:031905. [PMID: 30456343 PMCID: PMC6238644 DOI: 10.1063/1.5023503] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 05/31/2018] [Indexed: 11/14/2022] Open
Abstract
Glioblastoma (GBM), a highly aggressive form of brain tumor, is a disease marked by extensive invasion into the surrounding brain. Interstitial fluid flow (IFF), or the movement of fluid within the spaces between cells, has been linked to increased invasion of GBM cells. Better characterization of IFF could elucidate underlying mechanisms driving this invasion in vivo. Here, we develop a technique to noninvasively measure interstitial flow velocities in the glioma microenvironment of mice using dynamic contrast-enhanced magnetic resonance imaging (MRI), a common clinical technique. Using our in vitro model as a phantom "tumor" system and in silico models of velocity vector fields, we show we can measure average velocities and accurately reconstruct velocity directions. With our combined MR and analysis method, we show that velocity magnitudes are similar across four human GBM cell line xenograft models and the direction of fluid flow is heterogeneous within and around the tumors, and not always in the outward direction. These values were not linked to the tumor size. Finally, we compare our flow velocity magnitudes and the direction of flow to a classical marker of vessel leakage and bulk fluid drainage, Evans blue. With these data, we validate its use as a marker of high and low IFF rates and IFF in the outward direction from the tumor border in implanted glioma models. These methods show, for the first time, the nature of interstitial fluid flow in models of glioma using a technique that is translatable to clinical and preclinical models currently using contrast-enhanced MRI.
Collapse
Affiliation(s)
- Kathryn M. Kingsmore
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, Virginia 22904, USA
| | - Andrea Vaccari
- Department of Electrical and Computer Engineering, University of Virginia School of Engineering and Applied Science, Charlottesville, Virginia 22904, USA
| | - Daniel Abler
- Division of Mathematical Oncology, City of Hope, Duarte, California 91010, USA
| | - Sophia X. Cui
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, Virginia 22904, USA
| | - Frederick H. Epstein
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, Virginia 22904, USA
| | - Russell C. Rockne
- Division of Mathematical Oncology, City of Hope, Duarte, California 91010, USA
| | - Scott T. Acton
- Department of Electrical and Computer Engineering, University of Virginia School of Engineering and Applied Science, Charlottesville, Virginia 22904, USA
| | - Jennifer M. Munson
- Author to whom correspondence should be addressed: . Tel.: (540)-231-7896
| |
Collapse
|
27
|
Harris AR, Perez MJ, Munson JM. Docetaxel facilitates lymphatic-tumor crosstalk to promote lymphangiogenesis and cancer progression. BMC Cancer 2018; 18:718. [PMID: 29976154 PMCID: PMC6034223 DOI: 10.1186/s12885-018-4619-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 06/20/2018] [Indexed: 12/20/2022] Open
Abstract
Background Infiltration into lymphatic vessels is a critical step in breast cancer metastasis. Lymphatics undergo changes that facilitate metastasis as a result of activation of the cells lining lymphatic vessels, lymphatic endothelial cells (LECs). Inhibition of activation by targeting VEGFR3 can reduce invasion toward lymphatics. To best benefit patients, this approach should be coupled with standard of care that slows tumor growth, such as chemotherapy. Little is known about how chemotherapies, like docetaxel, may influence lymphatics and conversely, how lymphatics can alter responses to therapy. Methods A novel 3D in vitro co-culture model of the human breast tumor microenvironment was employed to examine the contribution of LECs to tumor invasion and viability with docetaxel and anti-VEGFR3, using three cell lines, MDA-MB-231, HCC38, and HCC1806. In vivo, the 4T1 mouse model of breast carcinoma was used to examine the efficacy of combinatorial therapy with docetaxel and anti-VEGFR3 on lymph node metastasis and tumor growth. Lymphangiogenesis in these mice was analyzed by immunohistochemistry and flow cytometry. Luminex analysis was used to measure expression of lymphangiogenic cytokines. Results In vitro, tumor cell invasion significantly increased with docetaxel when LECs were present; this effect was attenuated by inhibition of VEGFR3. LECs reduced docetaxel-induced cell death independent of VEGFR3. In vivo, docetaxel significantly increased breast cancer metastasis to the lymph node. Docetaxel and anti-VEGFR3 combination therapy reduced lymph node and lung metastasis in 4T1 and synergized to reduce tumor growth. Docetaxel induced VEGFR3-dependent vessel enlargement, lymphangiogenesis, and expansion of the LEC population in the peritumoral microenvironment, but not tumor-free stroma. Docetaxel caused an upregulation in pro-lymphangiogenic factors including VEGFC and TNF-α in the tumor microenvironment in vivo. Conclusions Here we present a counter-therapeutic effect of docetaxel chemotherapy that triggers cancer cells to elicit lymphangiogenesis. In turn, lymphatics reduce cancer response to docetaxel by altering the cytokine milieu in breast cancer. These changes lead to an increase in tumor cell invasion and survival under docetaxel treatment, ultimately reducing docetaxel efficacy. These docetaxel-induced effects can be mitigated by anti-VEGFR3 therapy, resulting in a synergism between these treatments that reduces tumor growth and metastasis. Electronic supplementary material The online version of this article (10.1186/s12885-018-4619-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alexandra R Harris
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Matthew J Perez
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA
| | - Jennifer M Munson
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA. .,Department of Biomedical Engineering & Mechanics, Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Virginia Polytechnic Institute & State University, Blacksburg, VA, 24061, USA.
| |
Collapse
|
28
|
Nguyen J, Jiao J, Smoot K, Watt GP, Zhao C, Song X, Stevenson HL, McCormick JB, Fisher-Hoch SP, Zhang J, Futreal PA, Beretta L. Toll-like receptor 4: a target for chemoprevention of hepatocellular carcinoma in obesity and steatohepatitis. Oncotarget 2018; 9:29495-29507. [PMID: 30034633 PMCID: PMC6047684 DOI: 10.18632/oncotarget.25685] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 06/13/2018] [Indexed: 02/06/2023] Open
Abstract
The incidence of hepatocellular carcinoma (HCC) associated with non-alcoholic fatty liver disease (NAFLD) is rapidly increasing. We aimed to elucidate the genetic basis of NAFLD-associated HCC and identify candidate targets for chemoprevention. Twenty HCC tumors, distant liver and matched tails from mice with hepatocyte-deletion of Pten (HepPten-) were subjected to whole-exome sequencing. A total of 162 genes with somatic non-synonymous single nucleotide variants or exonic small insertions and deletions in tumors were identified. Ingenuity Pathway Analysis of these 162 genes, further identified Toll-like receptor (TLR) 4, a key mediator of proinflammatory responses, and resatorvid, a TLR4 inhibitor, as the main causal networks of this dataset. Resatorvid treatment strongly prevented HCC development in these mice (p < 0.001). Remarkably, HCC patients with high tumoral TLR4 mRNA expression were more likely to be diagnosed with NAFLD and obese. TLR4 mRNA expression positively correlated with IL-6 and IL-10 mRNA expression in HCC tumors and the correlation was stronger in obese HCC patients. We have identified tumor mutation signatures and associated causal networks in NAFLD-associated HCC in HepPten- mice and further demonstrated the important role of TLR4 in promoting HCC development. This study also identified IL-6 and IL-10 as markers of TLR4 activation in HCC and subjects with NAFLD and obesity as the target population who would benefit from TLR4 inhibition treatment for HCC chemoprevention.
Collapse
Affiliation(s)
- Jennifer Nguyen
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jingjing Jiao
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kristin Smoot
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gordon P Watt
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,University of Texas Health Science Center at Houston, School of Public Health in Brownsville, Brownsville, TX, USA
| | - Chen Zhao
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xingzhi Song
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heather L Stevenson
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Joseph B McCormick
- University of Texas Health Science Center at Houston, School of Public Health in Brownsville, Brownsville, TX, USA
| | - Susan P Fisher-Hoch
- University of Texas Health Science Center at Houston, School of Public Health in Brownsville, Brownsville, TX, USA
| | - Jianhua Zhang
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - P Andrew Futreal
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Laura Beretta
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
29
|
Sun J, Luo Q, Liu L, Song G. Low-level shear stress promotes migration of liver cancer stem cells via the FAK-ERK1/2 signalling pathway. Cancer Lett 2018; 427:1-8. [PMID: 29678550 DOI: 10.1016/j.canlet.2018.04.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 03/26/2018] [Accepted: 04/12/2018] [Indexed: 10/24/2022]
Abstract
Cancer stem cells (CSCs) are a small subpopulation of tumour cells that have been proposed to be responsible for cancer initiation, chemotherapy resistance and cancer recurrence. Shear stress activated cellular signalling is involved in cellular migration, proliferation and differentiation. However, little is known about the effects of shear stress on the migration of liver cancer stem cells (LCSCs). Here, we studied the effects of shear stress that are generated from a parallel plated flow chamber system, on LCSC migration and the activation of focal adhesion kinase (FAK) and extracellular signal regulated kinase1/2 (ERK1/2), using transwell assay and western blot, respectively. We found that 2 dyne/cm2 shear stress loading for 6 h promotes LCSC migration and activation of the FAK and ERK1/2 signalling pathways, whereas treatment with the FAK phosphorylation inhibitor PF573228 or the ERK1/2 phosphorylation inhibitor PD98059 suppressed the shear stress-promoted migration, indicating the involvement of FAK and ERK1/2 activation in shear stress-induced LCSC migration. Additionally, atomic force microscopy (AFM) analysis showed that shear stress lowers LCSC stiffness via the FAK and ERK1/2 pathways, suggesting that the mechanism by which shear stress promotes LCSC migration might partially be responsible for the decrease in cell stiffness. Further experiments focused on the role of the actin cytoskeleton, demonstrating that the F-actin filaments in LCSCs are less well-defined after shear stress treatment, providing an explanation for the reduction in cell stiffness and the promotion of cell migration. Overall, our study demonstrates that shear stress promotes LCSC migration through the activation of the FAK-ERK1/2 signalling pathways, which further results in a reduction of organized actin and softer cell bodies.
Collapse
Affiliation(s)
- Jinghui Sun
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030, People's Republic of China; School of Medical Laboratory Science, Chengdu Medical College, Chengdu, 610500, People's Republic of China
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030, People's Republic of China
| | - Lingling Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030, People's Republic of China; School of Medical Laboratory Science, Chengdu Medical College, Chengdu, 610500, People's Republic of China
| | - Guanbin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030, People's Republic of China.
| |
Collapse
|
30
|
Fan W, Ye G. Microarray analysis for the identification of specific proteins and functional modules involved in the process of hepatocellular carcinoma originating from cirrhotic liver. Mol Med Rep 2018; 17:5619-5626. [PMID: 29436633 PMCID: PMC5866002 DOI: 10.3892/mmr.2018.8555] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 06/30/2017] [Indexed: 02/06/2023] Open
Abstract
In order to identify the potential pathogenesis of hepatocellular carcinoma (HCC) developing from cirrhosis, a microarray‑based transcriptome profile was analyzed. The GSE63898 expression profile was downloaded from the Gene Expression Omnibus database, which included data from 228 HCC tissue samples and 168 cirrhotic tissue samples. The Robust Multi‑array Average in the Affy package of R was used for raw data processing and Student's t‑test was used to screen differentially expressed genes (DEGs). An enrichment analysis was then conducted using the Database for Annotation, Visualization and Integrated Discovery online tool, and the protein‑protein interaction (PPI) network was constructed using the Search Tool for the Retrieval of Interacting Genes and Cytoscape. Furthermore, the MCODE plug‑in of Cytoscape was used to conduct a sub‑module analysis. A total of 634 DEGs were identified between HCC and cirrhosis, of which 165 were upregulated and 469 were downregulated. According to the cut‑off criteria, the PPI network was constructed and Jun proto‑oncogene, AP‑1 transcription factor subunit (degree, 39), Fos proto‑oncogene, AP‑1 transcription factor subunit (degree, 34) and v‑myc avian myelocytomatosis viral oncogene homolog (degree, 32) were identified as the hub nodes of the PPI network. Based on the sub‑module analysis, four specific modules were identified. In particular, module 1 was significantly enriched in the chemokine signaling pathway, and C‑X‑C motif chemokine ligand 12, C‑C motif chemokine receptor 7 (CCR7) and C‑C motif chemokine ligand 5 (CCL5) were three important proteins in this module. Module 4 was significantly enriched in chemical carcinogenesis, and cytochrome P450 family 2 subfamily E member 1, cytochrome P450 family 2 subfamily C member 9 (CYP2C9) and cytochrome P450 family 2 subfamily A member 6 (CYP2A6) were three important proteins in this module. In conclusion, the present study revealed that CCR7, CCL5, CYP2C9 and CYP2A6 are novel genes identified in the development of HCC; however, the actual functions of these genes require verification.
Collapse
Affiliation(s)
- Wufeng Fan
- Section of Medical Affairs, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei 441000, P.R. China
| | - Guangming Ye
- Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
31
|
Ghosh D, Dawson MR. Microenvironment Influences Cancer Cell Mechanics from Tumor Growth to Metastasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1092:69-90. [PMID: 30368749 DOI: 10.1007/978-3-319-95294-9_5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The microenvironment in a solid tumor includes a multitude of cell types, matrix proteins, and growth factors that profoundly influence cancer cell mechanics by providing both physical and chemical stimulation. This tumor microenvironment, which is both dynamic and heterogeneous in nature, plays a critical role in cancer progression from the growth of the primary tumor to the development of metastatic and drug-resistant tumors. This chapter provides an overview of the biophysical tools used to study cancer cell mechanics and mechanical changes in the tumor microenvironment at different stages of cancer progression, including growth of the primary tumor, local invasion, and metastasis. Quantitative single cell biophysical analysis of intracellular mechanics, cell traction forces, and cell motility can easily be combined with analysis of critical cell fate processes, including adhesion, proliferation, and drug resistance, to determine how changes in mechanics contribute to cancer progression. This biophysical approach can be used to systematically investigate the parameters in the tumor that control cancer cell interactions with the stroma and to identify specific conditions that induce tumor-promoting behavior, along with strategies for inhibiting these conditions to treat cancer. Increased understanding of the underlying biophysical mechanisms that drive cancer progression may provide insight into novel therapeutic approaches in the fight against cancer.
Collapse
Affiliation(s)
- Deepraj Ghosh
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI, USA
| | - Michelle R Dawson
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI, USA.
- Center for Biomedical Engineering, Brown University, Providence, RI, USA.
- School of Engineering, Brown University, Providence, RI, USA.
| |
Collapse
|
32
|
Fine N, Dimitriou ID, Rottapel R. Go with the flow: GEF-H1 mediated shear stress mechanotransduction in neutrophils. Small GTPases 2017; 11:23-31. [PMID: 29188751 DOI: 10.1080/21541248.2017.1332505] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Neutrophils in circulation experience significant shear forces due to blood flow when they tether to the vascular endothelium. Biochemical and biophysical responses of neutrophils to the physical force of flowing blood modulate their behavior and promote tissue recruitment under pro-inflammatory conditions. Neutrophil mechanotransduction responses occur through mechanisms that are not yet fully understood. In our recent work, we showed that GEF-H1, a RhoA specific guanine nucleotide exchange factor (GEF), is required to maintain neutrophil motility and migration in response to shear stress. GEF-H1 re-localizes to flottilin-rich uropods in neutrophils in response to fluid shear stress and promotes spreading and crawling on activated endothelial cells. GEF-H1 drives cellular contractility through myosin light chain (MLC) phosphorylation downstream of the Rho-ROCK signaling axis. We propose that GEF-H1-dependent cell spreading and crawling in shear stress-dependent neutrophil recruitment from the vasculature are due to the specific localization of Rho-induced contractility in the uropod.
Collapse
Affiliation(s)
- Noah Fine
- Matrix Dynamics Group, University of Toronto, Toronto, Ontario, Canada
| | - Ioannis D Dimitriou
- Princess Margaret Cancer Center, Toronto Medical Discovery Tower, Toronto, Ontario, Canada
| | - Robert Rottapel
- Princess Margaret Cancer Center, Toronto Medical Discovery Tower, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
33
|
Jeng KS, Jeng CJ, Jeng WJ, Chang CF, Sheen IS. Role of C-X-C chemokine ligand 12/C-X-C chemokine receptor 4 in the progression of hepatocellular carcinoma. Oncol Lett 2017; 14:1905-1910. [PMID: 28789425 DOI: 10.3892/ol.2017.6396] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 03/23/2017] [Indexed: 12/14/2022] Open
Abstract
The efficacy of the current non-surgical treatments for advanced hepatocellular carcinoma (HCC) remains limited and novel treatments are required to improve patient outcomes. The majority of HCCs develop from chronically damaged tissue that contains a high degree of inflammation and fibrosis, which promotes tumor progression and resistance to therapy. Understanding the interaction between stromal components and cancer cells (and the signaling pathways involved in this interaction) could aid the identification of novel therapeutic targets. Numerous studies have demonstrated a marked association between high C-X-C chemokine receptor 4 (CXCR4) expression and the invasiveness, progression and metastasis of HCC. The present review will investigate the different roles of CXCR4 in the progression of HCC and discuss possible future treatments. Through the C-X-C chemokine ligand 12 (CXCL12)/CXCR4 signaling pathway, ephrin A1 activation enhances the migration of endothelial progenitor cells to HCC to enable the neovascularization of tumors. There is an association between nuclear CXCR4 expression and the lymph node metastasis of HCC to distant areas. CXCR4 enhances cell migration in vitro and cell homing in vivo. CXCR4 levels are concentrated at the border of a tumor and in perivascular areas, inducing invasive behavior. The binding of CXCL12 to CXCR4 activates intracellular signaling pathways and induces crosstalk with transforming growth factor-β signaling, which enhances the migration of cancer cells. The CXCL12/CXCR4 axis also activates expression of matrix metalloproteinase 10, which further stimulates migration. CXCR4 is likely to crosstalk with the sonic hedgehog signaling pathway, contributing to tumor invasiveness and supporting the cancer stem-cell population; as a result, CXCR4 can be regarded as a cancer stem-cell marker. CXCR4 influences interstitial fluid flow-induced invasion. CXCR4 expression and HCC cell migration are promoted by α-fetoprotein, which activates AKT/mechanistic target of rapamycin signaling. CXCR4 also has the potential to affect sorafenib treatment for HCC. Targeting the CXCL12/CXCR4 signaling pathway may, therefore, be a promising strategy in HCC treatment.
Collapse
Affiliation(s)
- Kuo-Shyang Jeng
- Department of Surgery, Far Eastern Memorial Hospital, New Taipei 220, Taiwan, R.O.C.,Department of Medical Research, Far Eastern Memorial Hospital, New Taipei 220, Taiwan, R.O.C
| | - Chi-Juei Jeng
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 10048, Taiwan, R.O.C
| | - Wen-Juei Jeng
- Department of Hepato-Gastroenterology, Chang Gung Memorial Hospital Lin Kau Medical Center, Chang Gung University, Taoyuan 33, Taiwan, R.O.C
| | - Chiung-Fang Chang
- Department of Surgery, Far Eastern Memorial Hospital, New Taipei 220, Taiwan, R.O.C.,Department of Medical Research, Far Eastern Memorial Hospital, New Taipei 220, Taiwan, R.O.C
| | - I-Shyan Sheen
- Department of Hepato-Gastroenterology, Chang Gung Memorial Hospital Lin Kau Medical Center, Chang Gung University, Taoyuan 33, Taiwan, R.O.C
| |
Collapse
|
34
|
Ren T, Zhu L, Cheng M. CXCL10 accelerates EMT and metastasis by MMP-2 in hepatocellular carcinoma. Am J Transl Res 2017; 9:2824-2837. [PMID: 28670372 PMCID: PMC5489884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Accepted: 03/22/2017] [Indexed: 06/07/2023]
Abstract
Human malignant hepatocellular carcinoma (HCC) is a common tumor, which severely threatens human health and shortens longevity. The poor prognosis of HCC is primarily attributed to distant metastases. C-X-C motif chemokine 10 (CXCL10) regulates the control of several cellular and developmental processes including tumor cell proliferation, apoptosis, and cell metastasis. Previous studies have confirmed that CXCL10 functions as an oncogene in several cancers. However, the expression and biological functions of CXCL10 in HCC, especially with regard to metastasis, need further investigation. In this study, CXCL10 was found to be over expressed in invasive HCC cells and HCC clinical samples. While the over-expression of CXCL10 enhanced migration, invasion, and metastasis of HCC cells in vitro as well as in vivo, silencing of CXCL10 resulted in inhibition of HCC cell metastasis. Further, CXCL10 was found to accelerate epithelial-mesenchymal transition of HCC cells. The microarray analysis indicated that matrix metallopeptidase-2 (MMP-2) functions as a downstream factor of CXCL10. This study demonstrates that CXCL10 partakes in the metastasis of HCC by activating MMP-2 expression.
Collapse
Affiliation(s)
- Tingting Ren
- Department of Biochemistry, Affiliated Hospital of Guiyang Medical College28 Guiyi Street, Guiyang 550004, Guizhou, China
| | - Lili Zhu
- The Affiliated Baiyun Hospital of Guizhou Medical UniversityGuiyang 550004, Guizhou, China
| | - Mingliang Cheng
- Department of Infectious Diseases, Affiliated Hospital of Guiyang Medical College28 Guiyi Street, Guiyang 550004, Guizhou, China
| |
Collapse
|
35
|
Li Y, Yang X, Wu Y, Zhao K, Ye Z, Zhu J, Xu X, Zhao X, Xing C. B7-H3 promotes gastric cancer cell migration and invasion. Oncotarget 2017; 8:71725-71735. [PMID: 29069741 PMCID: PMC5641084 DOI: 10.18632/oncotarget.17847] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 04/27/2017] [Indexed: 02/06/2023] Open
Abstract
B7-H3 (B7 homologue 3, CD276) is a member of the B7 immunoregulatory family and promotes tumor progression. The present study demonstrated that B7-H3 promotes gastric cancer cell migration and invasion. shRNA-mediated B7-H3 silencing in the N87 gastric cancer cell line suppressed cell migration and invasion in vitro and in vivo; downregulated metastasis-associated CXCR4; and inhibited AKT, ERK, and Jak2/Stat3 phosphorylation. B7-H3-silenced cells injected into the tail veins of 4-week-old female BALB/c nude mice produced fewer metastases than control cells, and resulted in longer survival times. Immunofluorescence analyses confirmed B7-H3/CXCR4 colocalization in N87 cells, and co-immunoprecipitation assays showed a direct interaction between the two proteins. Our analysis of 120 tissue samples from gastric cancer patients showed that increased B7-H3 expression correlated positively with both tumor infiltration depth and CXCR4 expression. These findings suggest that B7-H3 and CXCR4 may be novel targets for anti-gastric cancer therapeutics.
Collapse
Affiliation(s)
- Yecheng Li
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu, P. R. China
| | - Xiaodong Yang
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu, P. R. China
| | - Yong Wu
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu, P. R. China
| | - Kui Zhao
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu, P. R. China
| | - Zhenyu Ye
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu, P. R. China
| | - Junjia Zhu
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu, P. R. China
| | - Xiaohui Xu
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu, P. R. China
| | - Xin Zhao
- Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou, 215006, P.R. China
| | - Chungen Xing
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu, P. R. China
| |
Collapse
|
36
|
Kingsmore KM, Logsdon DK, Floyd DH, Peirce SM, Purow BW, Munson JM. Interstitial flow differentially increases patient-derived glioblastoma stem cell invasionviaCXCR4, CXCL12, and CD44-mediated mechanisms. Integr Biol (Camb) 2016; 8:1246-1260. [DOI: 10.1039/c6ib00167j] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Kathryn M. Kingsmore
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Daniel K. Logsdon
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Desiree H. Floyd
- Department of Neurology, University of Virginia School of Medicine, Charlottesville, VA, 22908 USA
| | - Shayn M. Peirce
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Benjamin W. Purow
- Department of Neurology, University of Virginia School of Medicine, Charlottesville, VA, 22908 USA
| | - Jennifer M. Munson
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| |
Collapse
|
37
|
The role of polymorphisms of stromal-derived factor-1 and CXC receptor 4 in acute myeloid leukemia and leukemia cell dissemination. Gene 2016; 588:103-8. [PMID: 27154815 DOI: 10.1016/j.gene.2016.04.059] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 04/10/2016] [Accepted: 04/29/2016] [Indexed: 02/05/2023]
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a form of cancer characterized by infiltration of the bone marrow, blood, and other tissues by proliferative, clonal, abnormally differentiated cells of the hematopoietic system. Chemokine stromal cell-derived factor 1 (SDF-1) and its receptor CXC receptor 4 (CXCR4) play crucial roles in malignant cell invasion. Genetic polymorphisms may contribute to the differences in the expression level and activities associated with the SDF-1/CXCR4 pathway. This study aimed to determine the associations between the polymorphisms located on the SDF-1 (rs1801157, G>A) and CXCR4 (rs2228014, C>T) encoding genes and susceptibility and leukemia cell dissemination in AML. METHODS A total of 926 individuals, including 466 de novo AML patients and 460 healthy controls were genotyped for rs1801157 and rs2228014 using DNA Sanger sequencing. RESULTS Genotype distributions of CT and CT+TT for rs2228014 were significantly increased in AML patients compared with healthy controls [OR: 1.36, p=0.04; OR: 1.34, p=0.04; respectively]. However, rs1801157 demonstrated no significant differences in genotype distributions and allele frequency between AML patients and healthy controls. For the two combined SNPs, there was no significant proportional difference between the wild type GG-CC genotypes and non-GG-CC genotypes in AML patients and healthy controls. Additionally, peripheral blood leukemia-cell (PBLC) count was not statistically influenced by the genotypes of either rs1801157 or rs2228014. CONCLUSION Genotype CT of rs2228014 appeared to correlate with AML risk, but played no role in leukemia cells invading the bloodstream, while rs1801157 and the two combined SNPs were not associated with either increased AML risk or extramedullary leukemia-cell dissemination.
Collapse
|