1
|
Wan Z, Zhang S, Zhong AX, Xu L, Coughlin MF, Pavlou G, Shelton SE, Nguyen HT, Hirose S, Kim S, Floryan MA, Barbie DA, Hodi FS, Kamm RD. Transmural Flow Upregulates PD-L1 Expression in Microvascular Networks. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400921. [PMID: 38696611 PMCID: PMC11234398 DOI: 10.1002/advs.202400921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 03/24/2024] [Indexed: 05/04/2024]
Abstract
Endothelial programmed death-ligand 1 (PD-L1) expression is higher in tumors than in normal tissues. Also, tumoral vasculatures tend to be leakier than normal vessels leading to a higher trans-endothelial or transmural fluid flow. However, it is not clear whether such elevated transmural flow can control endothelial PD-L1 expression. Here, a new microfluidic device is developed to investigate the relationship between transmural flow and PD-L1 expression in microvascular networks (MVNs). After treating the MVNs with transmural flow for 24 h, the expression of PD-L1 in endothelial cells is upregulated. Additionally, CD8 T cell activation by phytohemagglutinin (PHA) is suppressed when cultured in the MVNs pre-conditioned with transmural flow. Moreover, transmural flow is able to further increase PD-L1 expression in the vessels formed in the tumor microenvironment. Finally, by utilizing blocking antibodies and knock-out assays, it is found that transmural flow-driven PD-L1 upregulation is controlled by integrin αVβ3. Overall, this study provides a new biophysical explanation for high PD-L1 expression in tumoral vasculatures.
Collapse
Affiliation(s)
- Zhengpeng Wan
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Shun Zhang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Amy X Zhong
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Liling Xu
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | - Mark F Coughlin
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Georgios Pavlou
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Sarah E Shelton
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Huu Tuan Nguyen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Satomi Hirose
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Seunggyu Kim
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Marie A Floryan
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - David A Barbie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Parker Institute for Cancer Immunotherapy, Boston, MA, 02215, USA
| | - Roger D Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
2
|
Zeng H, Li D, Dong J, Zhou X, Ou M, Xue W, Zhang R, Zou Y, Tang D, Yin L, Dai Y. Qualitative Proteome-wide Lysine Crotonylation Profiling Reveals Protein Modification Alteration in the Leukocyte Extravasation Pathway in Systemic Lupus Erythematosus. ACS OMEGA 2023; 8:44905-44919. [PMID: 38046296 PMCID: PMC10688171 DOI: 10.1021/acsomega.3c06293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/29/2023] [Accepted: 10/10/2023] [Indexed: 12/05/2023]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a severe systemic autoimmune disease with multiple manifestations. Lysine crotonylation (Kcr) is a newly discovered posttranslational modification epigenetic pattern that may affect gene expression and is linked to diseases causally. METHODS We collected blood samples from 11 SLE individuals and 36 healthy subjects. Then, we used highly sensitive liquid chromatography-mass spectrometry technology to carry out proteomics and quantitative crotonylome analysis of SLE peripheral blood mononuclear cells in this investigation, which indicated the unique etiology of SLE. Finally, we verified the expression of critical protein in the leukocyte extravasation pathway by online database analysis and Western blot. RESULTS There were 618 differentially expressed proteins (DEPs), and 612 crotonylated lysine sites for 272 differentially modified proteins (DMPs) found. These DEPs and DMPs are primarily enriched in the leukocyte extravasation signaling pathway, such as MMP8, MMP9, and ITGAM. CONCLUSIONS This is the first study of crotonylated modification proteomics in SLE. The leukocyte extravasation signaling pathway had a considerable concentration of DEPs and DMPs, indicating that this pathway may be involved in the pathogenic development of SLE.
Collapse
Affiliation(s)
- Huiyi Zeng
- Institute
of Nephrology and Blood Purification, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510632, China
- The First
Affiliated Hospital, School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, China
| | - Dandan Li
- Department
of Clinical Medical Research Center, The
Second Clinical Medical College of Jinan University, Shenzhen People’s
Hospital, Shenzhen, Guangdong 518020, China
- Experimental
Center, Shenzhen Pingle Orthopedic Hospital
(Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen, Guangdong 518118, China
- The First
Affiliated Hospital, School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, China
| | - Jingjing Dong
- Institute
of Nephrology and Blood Purification, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510632, China
- Department
of Clinical Medical Research Center, The
Second Clinical Medical College of Jinan University, Shenzhen People’s
Hospital, Shenzhen, Guangdong 518020, China
| | - Xianqing Zhou
- Guangxi
Key Laboratory of Metabolic Diseases Research, No. 924 Hospital of PLA Joint Logistic Support
Force, Guilin, Guangxi 541002, China
| | - Minglin Ou
- Guangxi
Key Laboratory of Metabolic Diseases Research, No. 924 Hospital of PLA Joint Logistic Support
Force, Guilin, Guangxi 541002, China
| | - Wen Xue
- Guangxi
Key Laboratory of Metabolic Diseases Research, No. 924 Hospital of PLA Joint Logistic Support
Force, Guilin, Guangxi 541002, China
| | - Ruohan Zhang
- Guangxi
Key Laboratory of Metabolic Diseases Research, No. 924 Hospital of PLA Joint Logistic Support
Force, Guilin, Guangxi 541002, China
| | - Yaoshuang Zou
- Guangxi
Key Laboratory of Metabolic Diseases Research, No. 924 Hospital of PLA Joint Logistic Support
Force, Guilin, Guangxi 541002, China
| | - Donge Tang
- Department
of Clinical Medical Research Center, The
Second Clinical Medical College of Jinan University, Shenzhen People’s
Hospital, Shenzhen, Guangdong 518020, China
| | - Lianghong Yin
- Institute
of Nephrology and Blood Purification, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510632, China
- Huangpu
Institute of Materials, Guangzhou, Guangdong 510663, China
| | - Yong Dai
- Department
of Clinical Medical Research Center, The
Second Clinical Medical College of Jinan University, Shenzhen People’s
Hospital, Shenzhen, Guangdong 518020, China
- Guangxi
Key Laboratory of Metabolic Diseases Research, No. 924 Hospital of PLA Joint Logistic Support
Force, Guilin, Guangxi 541002, China
- The First
Affiliated Hospital, School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, China
| |
Collapse
|
3
|
Shou Y, Teo XY, Wu KZ, Bai B, Kumar ARK, Low J, Le Z, Tay A. Dynamic Stimulations with Bioengineered Extracellular Matrix-Mimicking Hydrogels for Mechano Cell Reprogramming and Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300670. [PMID: 37119518 PMCID: PMC10375194 DOI: 10.1002/advs.202300670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/10/2023] [Indexed: 06/19/2023]
Abstract
Cells interact with their surrounding environment through a combination of static and dynamic mechanical signals that vary over stimulus types, intensity, space, and time. Compared to static mechanical signals such as stiffness, porosity, and topography, the current understanding on the effects of dynamic mechanical stimulations on cells remains limited, attributing to a lack of access to devices, the complexity of experimental set-up, and data interpretation. Yet, in the pursuit of emerging translational applications (e.g., cell manufacturing for clinical treatment), it is crucial to understand how cells respond to a variety of dynamic forces that are omnipresent in vivo so that they can be exploited to enhance manufacturing and therapeutic outcomes. With a rising appreciation of the extracellular matrix (ECM) as a key regulator of biofunctions, researchers have bioengineered a suite of ECM-mimicking hydrogels, which can be fine-tuned with spatiotemporal mechanical cues to model complex static and dynamic mechanical profiles. This review first discusses how mechanical stimuli may impact different cellular components and the various mechanobiology pathways involved. Then, how hydrogels can be designed to incorporate static and dynamic mechanical parameters to influence cell behaviors are described. The Scopus database is also used to analyze the relative strength in evidence, ranging from strong to weak, based on number of published literatures, associated citations, and treatment significance. Additionally, the impacts of static and dynamic mechanical stimulations on clinically relevant cell types including mesenchymal stem cells, fibroblasts, and immune cells, are evaluated. The aim is to draw attention to the paucity of studies on the effects of dynamic mechanical stimuli on cells, as well as to highlight the potential of using a cocktail of various types and intensities of mechanical stimulations to influence cell fates (similar to the concept of biochemical cocktail to direct cell fate). It is envisioned that this progress report will inspire more exciting translational development of mechanoresponsive hydrogels for biomedical applications.
Collapse
Affiliation(s)
- Yufeng Shou
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
- Institute for Health Innovation and TechnologyNational University of SingaporeSingapore117599Singapore
| | - Xin Yong Teo
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
| | - Kenny Zhuoran Wu
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
| | - Bingyu Bai
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
| | - Arun R. K. Kumar
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
- Institute for Health Innovation and TechnologyNational University of SingaporeSingapore117599Singapore
- Yong Loo Lin School of MedicineNational University of SingaporeSingapore117597Singapore
| | - Jessalyn Low
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
| | - Zhicheng Le
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
- Institute for Health Innovation and TechnologyNational University of SingaporeSingapore117599Singapore
| | - Andy Tay
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
- Institute for Health Innovation and TechnologyNational University of SingaporeSingapore117599Singapore
- NUS Tissue Engineering ProgramNational University of SingaporeSingapore117510Singapore
| |
Collapse
|
4
|
Rossi B, Dusi S, Angelini G, Bani A, Lopez N, Della Bianca V, Pietronigro EC, Zenaro E, Zocco C, Constantin G. Alpha4 beta7 integrin controls Th17 cell trafficking in the spinal cord leptomeninges during experimental autoimmune encephalomyelitis. Front Immunol 2023; 14:1071553. [PMID: 37143680 PMCID: PMC10151683 DOI: 10.3389/fimmu.2023.1071553] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 04/05/2023] [Indexed: 05/06/2023] Open
Abstract
Th1 and Th17 cell migration into the central nervous system (CNS) is a fundamental process in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), the animal model of multiple sclerosis (MS). Particularly, leptomeningeal vessels of the subarachnoid space (SAS) constitute a central route for T cell entry into the CNS during EAE. Once migrated into the SAS, T cells show an active motility behavior, which is a prerequisite for cell-cell communication, in situ reactivation and neuroinflammation. However, the molecular mechanisms selectively controlling Th1 and Th17 cell trafficking in the inflamed leptomeninges are not well understood. By using epifluorescence intravital microscopy, we obtained results showing that myelin-specific Th1 and Th17 cells have different intravascular adhesion capacity depending on the disease phase, with Th17 cells being more adhesive at disease peak. Inhibition of αLβ2 integrin selectively blocked Th1 cell adhesion, but had no effect on Th17 rolling and arrest capacity during all disease phases, suggesting that distinct adhesion mechanisms control the migration of key T cell populations involved in EAE induction. Blockade of α4 integrins affected myelin-specific Th1 cell rolling and arrest, but only selectively altered intravascular arrest of Th17 cells. Notably, selective α4β7 integrin blockade inhibited Th17 cell arrest without interfering with intravascular Th1 cell adhesion, suggesting that α4β7 integrin is predominantly involved in Th17 cell migration into the inflamed leptomeninges in EAE mice. Two-photon microscopy experiments showed that blockade of α4 integrin chain or α4β7 integrin selectively inhibited the locomotion of extravasated antigen-specific Th17 cells in the SAS, but had no effect on Th1 cell intratissue dynamics, further pointing to α4β7 integrin as key molecule in Th17 cell trafficking during EAE development. Finally, therapeutic inhibition of α4β7 integrin at disease onset by intrathecal injection of a blocking antibody attenuated clinical severity and reduced neuroinflammation, further demonstrating a crucial role for α4β7 integrin in driving Th17 cell-mediated disease pathogenesis. Altogether, our data suggest that a better knowledge of the molecular mechanisms controlling myelin-specific Th1 and Th17 cell trafficking during EAE delevopment may help to identify new therapeutic strategies for CNS inflammatory and demyelinating diseases.
Collapse
Affiliation(s)
- Barbara Rossi
- Department of Medicine, University of Verona, Verona, Italy
- *Correspondence: Barbara Rossi, ; Gabriela Constantin,
| | - Silvia Dusi
- Department of Medicine, University of Verona, Verona, Italy
| | | | | | - Nicola Lopez
- Department of Medicine, University of Verona, Verona, Italy
| | | | | | - Elena Zenaro
- Department of Medicine, University of Verona, Verona, Italy
| | - Carlotta Zocco
- Department of Medicine, University of Verona, Verona, Italy
| | - Gabriela Constantin
- Department of Medicine, University of Verona, Verona, Italy
- The Center for Biomedical Computing (CBMC), University of Verona, Verona, Italy
- *Correspondence: Barbara Rossi, ; Gabriela Constantin,
| |
Collapse
|
5
|
Kim SHJ, Hammer DA. Integrin cross-talk modulates stiffness-independent motility of CD4+ T lymphocytes. Mol Biol Cell 2021; 32:1749-1757. [PMID: 34232700 PMCID: PMC8684734 DOI: 10.1091/mbc.e21-03-0131] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
To carry out their physiological responsibilities, CD4+ T lymphocytes interact with various tissues of different mechanical properties. Recent studies suggest that T cells migrate upstream on surfaces expressing intracellular adhesion molecule-1 (ICAM-1) through interaction with leukocyte function-associated antigen-1 (αLβ2) (LFA-1) integrins. LFA-1 likely behaves as a mechanosensor, and thus we hypothesized that substrate mechanics might affect the ability of LFA-1 to support upstream migration of T cells under flow. Here we measured motility of CD4+ T lymphocytes on polyacrylamide gels with predetermined stiffnesses containing ICAM-1, vascular cell adhesion molecule-1 (VCAM-1), or a 1:1 mixture of VCAM-1/ICAM-1. Under static conditions, we found that CD4+ T cells exhibit an increase in motility on ICAM-1, but not on VCAM-1 or VCAM-1/ICAM-1 mixed, surfaces as a function of matrix stiffness. The mechanosensitivity of T-cell motility on ICAM-1 is overcome when VLA-4 (very late antigen-4 [α4β1]) is ligated with soluble VCAM-1. Last, we observed that CD4+ T cells migrate upstream under flow on ICAM-1-functionalized hydrogels, independent of substrate stiffness. In summary, we show that CD4+ T cells under no flow respond to matrix stiffness through LFA-1, and that the cross-talk of VLA-4 and LFA-1 can compensate for deformable substrates. Interestingly, CD4+ T lymphocytes migrated upstream on ICAM-1 regardless of the substrate stiffness, suggesting that flow can compensate for substrate stiffness.
Collapse
Affiliation(s)
- Sarah Hyun Ji Kim
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104
| | - Daniel A Hammer
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
6
|
Yazdian Kashani S, Keshavarz Moraveji M, Bonakdar S. Computational and experimental studies of a cell-imprinted-based integrated microfluidic device for biomedical applications. Sci Rep 2021; 11:12130. [PMID: 34108580 PMCID: PMC8190060 DOI: 10.1038/s41598-021-91616-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/27/2021] [Indexed: 02/05/2023] Open
Abstract
It has been proved that cell-imprinted substrates molded from template cells can be used for the re-culture of that cell while preserving its normal behavior or to differentiate the cultured stem cells into the template cell. In this study, a microfluidic device was presented to modify the previous irregular cell-imprinted substrate and increase imprinting efficiency by regular and objective cell culture. First, a cell-imprinted substrate from template cells was prepared using a microfluidic chip in a regular pattern. Another microfluidic chip with the same pattern was then aligned on the cell-imprinted substrate to create a chondrocyte-imprinted-based integrated microfluidic device. Computational fluid dynamics (CFD) simulations were used to obtain suitable conditions for injecting cells into the microfluidic chip before performing experimental evaluations. In this simulation, the effect of input flow rate, number per unit volume, and size of injected cells in two different chip sizes were examined on exerted shear stress and cell trajectories. This numerical simulation was first validated with experiments with cell lines. Finally, chondrocyte was used as template cell to evaluate the chondrogenic differentiation of adipose-derived mesenchymal stem cells (ADSCs) in the chondrocyte-imprinted-based integrated microfluidic device. ADSCs were positioned precisely on the chondrocyte patterns, and without using any chemical growth factor, their fibroblast-like morphology was modified to the spherical morphology of chondrocytes after 14 days of culture. Both immunostaining and gene expression analysis showed improvement in chondrogenic differentiation compared to traditional imprinting methods. This study demonstrated the effectiveness of cell-imprinted-based integrated microfluidic devices for biomedical applications.
Collapse
Affiliation(s)
- Sepideh Yazdian Kashani
- Department of Chemical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, 1591634311, Iran
| | - Mostafa Keshavarz Moraveji
- Department of Chemical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, 1591634311, Iran.
| | - Shahin Bonakdar
- National Cell Bank Department, Pasteur Institute of Iran, P.O. Box 13169-43551, Tehran, Iran.
| |
Collapse
|
7
|
Schwartz AB, Campos OA, Criado-Hidalgo E, Chien S, del Álamo JC, Lasheras JC, Yeh YT. Elucidating the Biomechanics of Leukocyte Transendothelial Migration by Quantitative Imaging. Front Cell Dev Biol 2021; 9:635263. [PMID: 33855018 PMCID: PMC8039384 DOI: 10.3389/fcell.2021.635263] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/09/2021] [Indexed: 01/13/2023] Open
Abstract
Leukocyte transendothelial migration is crucial for innate immunity and inflammation. Upon tissue damage or infection, leukocytes exit blood vessels by adhering to and probing vascular endothelial cells (VECs), breaching endothelial cell-cell junctions, and transmigrating across the endothelium. Transendothelial migration is a critical rate-limiting step in this process. Thus, leukocytes must quickly identify the most efficient route through VEC monolayers to facilitate a prompt innate immune response. Biomechanics play a decisive role in transendothelial migration, which involves intimate physical contact and force transmission between the leukocytes and the VECs. While quantifying these forces is still challenging, recent advances in imaging, microfabrication, and computation now make it possible to study how cellular forces regulate VEC monolayer integrity, enable efficient pathfinding, and drive leukocyte transmigration. Here we review these recent advances, paying particular attention to leukocyte adhesion to the VEC monolayer, leukocyte probing of endothelial barrier gaps, and transmigration itself. To offer a practical perspective, we will discuss the current views on how biomechanics govern these processes and the force microscopy technologies that have enabled their quantitative analysis, thus contributing to an improved understanding of leukocyte migration in inflammatory diseases.
Collapse
Affiliation(s)
- Amy B. Schwartz
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
| | - Obed A. Campos
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
| | - Ernesto Criado-Hidalgo
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Juan C. del Álamo
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
- Department of Mechanical Engineering, University of Washington, Seattle, WA, United States
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, United States
| | - Juan C. Lasheras
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Yi-Ting Yeh
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
8
|
Cheng X, Cheng K. Visualizing cancer extravasation: from mechanistic studies to drug development. Cancer Metastasis Rev 2021; 40:71-88. [PMID: 33156478 PMCID: PMC7897269 DOI: 10.1007/s10555-020-09942-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023]
Abstract
Metastasis is a multistep process that accounts for the majority of cancer-related death. By the end of metastasize dissemination, circulating tumor cells (CTC) need to extravasate the blood vessels at metastatic sites to form new colonization. Although cancer cell extravasation is a crucial step in cancer metastasis, it has not been successfully targeted by current anti-metastasis strategies due to the lack of a thorough understanding of the molecular mechanisms that regulate this process. This review focuses on recent progress in cancer extravasation visualization techniques, including the development of both in vitro and in vivo cancer extravasation models, that shed light on the underlying mechanisms. Specifically, multiple cancer extravasation stages, such as the adhesion to the endothelium and transendothelial migration, are successfully probed using these technologies. Moreover, the roles of different cell adhesive molecules, chemokines, and growth factors, as well as the mechanical factors in these stages are well illustrated. Deeper understandings of cancer extravasation mechanisms offer us new opportunities to escalate the discovery of anti-extravasation drugs and therapies and improve the prognosis of cancer patients.
Collapse
Affiliation(s)
- Xiao Cheng
- Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina at Chapel Hill, Raleigh, NC, USA
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, 27607, USA
| | - Ke Cheng
- Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina at Chapel Hill, Raleigh, NC, USA.
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, 27607, USA.
| |
Collapse
|
9
|
Le Master E, Ahn SJ, Levitan I. Mechanisms of endothelial stiffening in dyslipidemia and aging: Oxidized lipids and shear stress. CURRENT TOPICS IN MEMBRANES 2020; 86:185-215. [PMID: 33837693 PMCID: PMC8168803 DOI: 10.1016/bs.ctm.2020.08.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Vascular stiffening of the arterial walls is well-known as a key factor in aging and the development of cardiovascular disease; however, the role of endothelial stiffness in vascular dysfunction is still an emerging topic. In this review, the authors discuss the impact of dyslipidemia, oxidized lipids, substrate stiffness, age and pro-atherogenic disturbed flow have on endothelial stiffness. Furthermore, we investigate several mechanistic pathways that are key contributors in endothelial stiffness and discuss their physiological effects in the onset of atherogenesis in the disturbed flow regions of the aortic vasculature. The findings in this chapter describe a novel paradigm of synergistic interaction of plasma dyslipidemia/oxidized lipids and pro-atherogenic disturbed shear stress, as well as aging has on endothelial stiffness and vascular dysfunction.
Collapse
Affiliation(s)
- Elizabeth Le Master
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Sang Joon Ahn
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Irena Levitan
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Illinois at Chicago, Chicago, IL, United States.
| |
Collapse
|
10
|
Gordon E, Schimmel L, Frye M. The Importance of Mechanical Forces for in vitro Endothelial Cell Biology. Front Physiol 2020; 11:684. [PMID: 32625119 PMCID: PMC7314997 DOI: 10.3389/fphys.2020.00684] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/26/2020] [Indexed: 12/12/2022] Open
Abstract
Blood and lymphatic vessels are lined by endothelial cells which constantly interact with their luminal and abluminal extracellular environments. These interactions confer physical forces on the endothelium, such as shear stress, stretch and stiffness, to mediate biological responses. These physical forces are often altered during disease, driving abnormal endothelial cell behavior and pathology. Therefore, it is critical that we understand the mechanisms by which endothelial cells respond to physical forces. Traditionally, endothelial cells in culture are grown in the absence of flow on stiff substrates such as plastic or glass. These cells are not subjected to the physical forces that endothelial cells endure in vivo, thus the results of these experiments often do not mimic those observed in the body. The field of vascular biology now realize that an intricate analysis of endothelial signaling mechanisms requires complex in vitro systems to mimic in vivo conditions. Here, we will review what is known about the mechanical forces that guide endothelial cell behavior and then discuss the advancements in endothelial cell culture models designed to better mimic the in vivo vascular microenvironment. A wider application of these technologies will provide more biologically relevant information from cultured cells which will be reproducible to conditions found in the body.
Collapse
Affiliation(s)
- Emma Gordon
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Lilian Schimmel
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Maike Frye
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
11
|
Wettschureck N, Strilic B, Offermanns S. Passing the Vascular Barrier: Endothelial Signaling Processes Controlling Extravasation. Physiol Rev 2019; 99:1467-1525. [PMID: 31140373 DOI: 10.1152/physrev.00037.2018] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A central function of the vascular endothelium is to serve as a barrier between the blood and the surrounding tissue of the body. At the same time, solutes and cells have to pass the endothelium to leave or to enter the bloodstream to maintain homeostasis. Under pathological conditions, for example, inflammation, permeability for fluid and cells is largely increased in the affected area, thereby facilitating host defense. To appropriately function as a regulated permeability filter, the endothelium uses various mechanisms to allow solutes and cells to pass the endothelial layer. These include transcellular and paracellular pathways of which the latter requires remodeling of intercellular junctions for its regulation. This review provides an overview on endothelial barrier regulation and focuses on the endothelial signaling mechanisms controlling the opening and closing of paracellular pathways for solutes and cells such as leukocytes and metastasizing tumor cells.
Collapse
Affiliation(s)
- Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Boris Strilic
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| |
Collapse
|
12
|
Huselstein C, Rahouadj R, de Isla N, Bensoussan D, Stoltz JF, Li YP. Mechanobiology of mesenchymal stem cells: Which interest for cell-based treatment? Biomed Mater Eng 2017; 28:S47-S56. [PMID: 28372277 DOI: 10.3233/bme-171623] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Thanks to their immune properties, the mesenchymal stem cells (MSC) are a promising source for cell therapy. Current clinical trials show that MSC administrated to patients can treat different diseases (graft-versus-host disease (GVHD), liver cirrhosis, systemic lupus, erythematosus, rheumatoid arthritis, type I diabetes…). In this case, the most common mode of cell administration is the intravenous injection, and the hemodynamic environment of cells induced by blood circulation could interfere on their behavior during the migration and homing towards the injured site. After a brief review of the mechanobiology concept, this paper will help in understanding how the mechanical environment could interact with MSC behavior once they are injected to patient in cell-based treatment.
Collapse
Affiliation(s)
- Céline Huselstein
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle, 54500 Vandœuvre-lès-Nancy, France.,Université de Lorraine, 54000 Nancy, France.,FR3209 CNRS BMCT - Bio-Ingénierie Moléculaire Cellulaire et Thérapeutique, Faculté de Médecine, F-54505 Vandœuvre-lès-Nancy, France
| | - R Rahouadj
- Université de Lorraine, 54000 Nancy, France.,UMR 7563 CNRS-Université de Lorraine, LEMTA, Vandœuvre-lès-Nancy, France
| | - N de Isla
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle, 54500 Vandœuvre-lès-Nancy, France.,Université de Lorraine, 54000 Nancy, France.,FR3209 CNRS BMCT - Bio-Ingénierie Moléculaire Cellulaire et Thérapeutique, Faculté de Médecine, F-54505 Vandœuvre-lès-Nancy, France
| | - D Bensoussan
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle, 54500 Vandœuvre-lès-Nancy, France.,Université de Lorraine, 54000 Nancy, France.,FR3209 CNRS BMCT - Bio-Ingénierie Moléculaire Cellulaire et Thérapeutique, Faculté de Médecine, F-54505 Vandœuvre-lès-Nancy, France.,CHU de Nancy, Unité de Thérapie Cellulaire, banque de Tissus, 54500 Vandœuvre-lès-Nancy, France
| | - J F Stoltz
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle, 54500 Vandœuvre-lès-Nancy, France.,Université de Lorraine, 54000 Nancy, France.,FR3209 CNRS BMCT - Bio-Ingénierie Moléculaire Cellulaire et Thérapeutique, Faculté de Médecine, F-54505 Vandœuvre-lès-Nancy, France
| | - Y P Li
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| |
Collapse
|
13
|
The effect of substrate stiffness on cancer cell volume homeostasis. J Cell Physiol 2017; 233:1414-1423. [DOI: 10.1002/jcp.26026] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 05/22/2017] [Indexed: 12/30/2022]
|
14
|
BASAVARAJA PRASHANTH, SURENDRAN ANISH, GUPTA AJAY, SABA LUCA, LAIRD JOHNR, NICOLAIDES ANDREW, MTUI EDWARDE, BARADARAN HEDIYEH, LAVRA FRANCESCO, SURI JASJITS. WALL SHEAR STRESS AND OSCILLATORY SHEAR INDEX DISTRIBUTION IN CAROTID ARTERY WITH VARYING DEGREE OF STENOSIS: A HEMODYNAMIC STUDY. J MECH MED BIOL 2017. [DOI: 10.1142/s0219519417500373] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
A significant proportion of cerebral stroke is a consequence of the arterial stenotic plaque rupture causing local thrombosis or distal embolization. The formation and subsequent rupture of the plaque depends on wall shear stress (WSS) and oscillatory shear index (OSI). The purpose of the present study was to understand the effect of hemodynamics on the spatial and temporal variations of WSS and OSI using realistic models with varying degree of carotid artery stenosis (DOS). Multiple CT volumes were obtained from subjects in the carotid bifurcation zone and the 3D models were generated. A finite volume-based computational fluid dynamics (CFD) method was utilized to understand the hemodynamics in pulsatile flow conditions. It was observed that high stenosis models occupied a large value of normalized WSS in the internal carotid artery (ICA) whereas they had smaller values of normalized WSS in the common carotid artery (CCA). For clinical use, the authors recommend using the spatial average value of oscillatory shear rather than the maximum value for an accurate knowledge about the severity of stenosis. The resultant vorticity changes the direction of spin after the bifurcation zone. Additionally, we propose the use of limiting streamlines as a novel and convenient method to identify the disturbed flow regions that are prone to atherogenesis.
Collapse
Affiliation(s)
- PRASHANTH BASAVARAJA
- Department of Mechanical Engineering, National Institute of Technology Karnataka Surathkal, Mangalore 575025, Karnataka, India
| | - ANISH SURENDRAN
- Department of Mechanical Engineering, National Institute of Technology Karnataka Surathkal, Mangalore 575025, Karnataka, India
| | - AJAY GUPTA
- Department of Radiology, Weill Cornell Medical College, New York NY 10065, USA
| | - LUCA SABA
- Department of Radiology, Azienda Ospedaliero Universitaria (A.O.U.), di Cagliari — Polo di Monserrato, S. S. 554 Monserrato, Cagliari 09045, Italy
| | - JOHN R. LAIRD
- UC Davis Vascular Center, University of California Sacramento, CA 95817, USA
| | - ANDREW NICOLAIDES
- Vascular Screening and Diagnostic Centre, London W1G 6LF, UK
- Department of Biological Sciences, University of Cyprus, 1678 Nicosia, Cyprus
| | - EDWARD E. MTUI
- Department of Radiology, Weill Cornell Medical College, New York NY 10065, USA
| | - HEDIYEH BARADARAN
- Department of Radiology, Weill Cornell Medical College, New York NY 10065, USA
| | - FRANCESCO LAVRA
- Department of Radiology, Azienda Ospedaliero Universitaria (A.O.U.), di Cagliari — Polo di Monserrato, S. S. 554 Monserrato, Cagliari 09045, Italy
| | - JASJIT S. SURI
- Point of Care Devices, Global Biomedical Technologies, Inc., Roseville, CA 95661, USA
- Diagnostic and Monitoring Division, AtheroPointTM LLC, Roseville, CA 95661, USA
- Electrical Engineering Department, Idaho State University, Pocatello, ID 83209, USA
| |
Collapse
|
15
|
Choi JY, Jo SA. KDM7A histone demethylase mediates TNF-α-induced ICAM1 protein upregulation by modulating lysosomal activity. Biochem Biophys Res Commun 2016; 478:1355-62. [PMID: 27565733 DOI: 10.1016/j.bbrc.2016.08.128] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 08/22/2016] [Indexed: 11/24/2022]
Abstract
Intercellular adhesion molecule1 (ICAM1) is involved in adhesion and transmigration of leukocytes across endothelium, promoting brain inflammation and leading to brain diseases. Here, we studied the mechanism that regulates ICAM expression in response to proinflammatory cytokine tumor necrosis factor alpha (TNF-α). ICAM1 mRNA and protein levels in human brain microvascular endothelial cells dramatically increased after TNF-α treatment. TNF-α also upregulated histone demethylases KDM1B and KDM7A responsible for demethylation of H3K9me2, a well-known repression marker. Knockdown of KDM7A by a small interfering RNA reduced the ICAM1 protein level and leukocyte adhesion without an effect on ICAM1 mRNA expression. In contrast, a KDM1B knockdown did not affect TNF-α-induced ICAM1 expression. Thus, KDM7A-mediated ICAM1 protein upregulation is likely related to protein stability, not a histone-mediated epigenetic mechanism. Experiments with cycloheximide supported the role of KDM7A in ICAM1 protein stabilization. Further experiments suggest that KDM7A regulates ICAM1 protein stability via a lysosome-dependent pathway. Lysosome inhibitors increased the TNF-α-induced ICAM1 protein level and restored KDM7A knockdown-induced downregulation of ICAM1. In contrast, the KDM7A knockdown had no effect on proteasome-mediated ICAM1 degradation. We also found that the transcription factor EB protein level reduced in response to TNF-α but increased by the KDM7A knockdown. Immunocytochemical analysis revealed weak lysosome formation with high ICAM1 immunoreactivity after TNF-α treatment, but KDM7A knockdown reversed this response, resulting in strong lysosome formation with ICAM1 protein clustering in lysosomes. Taken together, our results show that KDM7A mediates TNF-α-induced ICAM1 protein upregulation and is mediated by induction of KDM7A, which regulates the TFEB-mediated lysosomal activity.
Collapse
Affiliation(s)
- Ji-Young Choi
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, South Korea
| | - Sangmee Ahn Jo
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, South Korea; Department of Pharmacology, College of Pharmacy, Dankook University, Cheonan, 31116, South Korea.
| |
Collapse
|
16
|
Schnoor M. Endothelial actin-binding proteins and actin dynamics in leukocyte transendothelial migration. THE JOURNAL OF IMMUNOLOGY 2015; 194:3535-41. [PMID: 25848070 DOI: 10.4049/jimmunol.1403250] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The endothelium is the first barrier that leukocytes have to overcome during recruitment to sites of inflamed tissues. The leukocyte extravasation cascade is a complex multistep process that requires the activation of various adhesion molecules and signaling pathways, as well as actin remodeling, in both leukocytes and endothelial cells. Endothelial adhesion molecules, such as E-selectin or ICAM-1, are connected to the actin cytoskeleton via actin-binding proteins (ABPs). Although the contribution of receptor-ligand interactions to leukocyte extravasation has been studied extensively, the contribution of endothelial ABPs to the regulation of leukocyte adhesion and transendothelial migration remains poorly understood. This review focuses on recently published evidence that endothelial ABPs, such as cortactin, myosin, or α-actinin, regulate leukocyte extravasation by controlling actin dynamics, biomechanical properties of endothelia, and signaling pathways, such as GTPase activation, during inflammation. Thus, ABPs may serve as targets for novel treatment strategies for disorders characterized by excessive leukocyte recruitment.
Collapse
Affiliation(s)
- Michael Schnoor
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, 07360 Mexico City, Mexico
| |
Collapse
|
17
|
Heemskerk N, van Rijssel J, van Buul JD. Rho-GTPase signaling in leukocyte extravasation: an endothelial point of view. Cell Adh Migr 2015; 8:67-75. [PMID: 24621576 PMCID: PMC4049863 DOI: 10.4161/cam.28244] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Leukocyte transendothelial migration (TEM) is one of the crucial steps during inflammation. A better understanding of the key molecules that regulate leukocyte extravasation aids to the development of novel therapeutics for treatment of inflammation-based diseases, such as atherosclerosis and rheumatoid arthritis. The adhesion molecules ICAM-1 and VCAM-1 are known as central mediators of TEM. Clustering of these molecules by their leukocytic integrins initiates the activation of several signaling pathways within the endothelium, including a rise in intracellular Ca (2+), activation of several kinase cascades, and the activation of Rho-GTPases. Activation of Rho-GTPases has been shown to control adhesion molecule clustering and the formation of apical membrane protrusions that embrace adherent leukocytes during TEM. Here, we discuss the potential regulatory mechanisms of leukocyte extravasation from an endothelial point of view, with specific focus on the role of the Rho-GTPases.
Collapse
Affiliation(s)
- Niels Heemskerk
- Department of Molecular Cell Biology; Sanquin Research and Landsteiner Laboratory; Academic Medical Center; University of Amsterdam; Amsterdam, the Netherlands
| | - Jos van Rijssel
- Department of Molecular Cell Biology; Sanquin Research and Landsteiner Laboratory; Academic Medical Center; University of Amsterdam; Amsterdam, the Netherlands
| | - Jaap D van Buul
- Department of Molecular Cell Biology; Sanquin Research and Landsteiner Laboratory; Academic Medical Center; University of Amsterdam; Amsterdam, the Netherlands
| |
Collapse
|
18
|
Oulaid O, Zhang J. Temporal and spatial variations of wall shear stress in the entrance region of microvessels. J Biomech Eng 2015; 137:061008. [PMID: 25781004 DOI: 10.1115/1.4030055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Indexed: 11/08/2022]
Abstract
Using a simplified two-dimensional divider-channel setup, we simulate the development process of red blood cell (RBC) flows in the entrance region of microvessels to study the wall shear stress (WSS) behaviors. Significant temporal and spatial variation in WSS is noticed. The maximum WSS magnitude and the strongest variation are observed at the channel inlet due to the close cell-wall contact. From the channel inlet, both the mean WSS and variation magnitude decrease, with a abrupt drop in the close vicinity near the inlet and then a slow relaxation over a relatively long distance; and a relative stable state with approximately constant mean and variation is established when the flow is well developed. The correlations between the WSS variation features and the cell free layer (CFL) structure are explored, and the effects of several hemodynamic parameters on the WSS variation are examined. In spite of the model limitations, the qualitative information revealed in this study could be useful for better understanding relevant processes and phenomena in the microcirculation.
Collapse
|
19
|
Marjoram RJ, Lessey EC, Burridge K. Regulation of RhoA activity by adhesion molecules and mechanotransduction. Curr Mol Med 2014; 14:199-208. [PMID: 24467208 PMCID: PMC3929014 DOI: 10.2174/1566524014666140128104541] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 07/05/2013] [Accepted: 12/02/2013] [Indexed: 12/26/2022]
Abstract
The low molecular weight GTP-binding protein RhoA regulates many cellular events, including cell migration, organization of the cytoskeleton, cell adhesion, progress through the cell cycle and gene expression. Physical forces influence these cellular processes in part by regulating RhoA activity through mechanotransduction of cell adhesion molecules (e.g. integrins, cadherins, Ig superfamily molecules). RhoA activity is regulated by guanine nucleotide exchange factors (GEFs) and GTPase activating proteins (GAPs) that are themselves regulated by many different signaling pathways. Significantly, the engagement of many cell adhesion molecules can affect RhoA activity in both positive and negative ways. In this brief review, we consider how RhoA activity is regulated downstream from cell adhesion molecules and mechanical force. Finally, we highlight the importance of mechanotransduction signaling to RhoA in normal cell biology as well as in certain pathological states.
Collapse
Affiliation(s)
| | | | - K Burridge
- Department of Cell Biology and Physiology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
20
|
Hamilla SM, Stroka KM, Aranda-Espinoza H. VE-cadherin-independent cancer cell incorporation into the vascular endothelium precedes transmigration. PLoS One 2014; 9:e109748. [PMID: 25275457 PMCID: PMC4183660 DOI: 10.1371/journal.pone.0109748] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 09/10/2014] [Indexed: 12/31/2022] Open
Abstract
Metastasis is accountable for 90% of cancer deaths. During metastasis, tumor cells break away from the primary tumor, enter the blood and the lymph vessels, and use them as highways to travel to distant sites in the body to form secondary tumors. Cancer cell migration through the endothelium and into the basement membrane represents a critical step in the metastatic cascade, yet it is not well understood. This process is well characterized for immune cells that routinely transmigrate through the endothelium to sites of infection, inflammation, or injury. Previous studies with leukocytes have demonstrated that this step depends heavily on the activation status of the endothelium and subendothelial substrate stiffness. Here, we used a previously established in vitro model of the endothelium and live cell imaging, in order to observe cancer cell transmigration and compare this process to leukocytes. Interestingly, cancer cell transmigration includes an additional step, which we term ‘incorporation’, into the endothelial cell (EC) monolayer. During this phase, cancer cells physically displace ECs, leading to the dislocation of EC VE-cadherin away from EC junctions bordering cancer cells, and spread into the monolayer. In some cases, ECs completely detach from the matrix. Furthermore, cancer cell incorporation occurs independently of the activation status and the subendothelial substrate stiffness for breast cancer and melanoma cells, a notable difference from the process by which leukocytes transmigrate. Meanwhile, pancreatic cancer cell incorporation was dependent on the activation status of the endothelium and changed on very stiff subendothelial substrates. Collectively, our results provide mechanistic insights into tumor cell extravasation and demonstrate that incorporation is one of the earliest steps.
Collapse
Affiliation(s)
- Susan M. Hamilla
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, United States of America
| | - Kimberly M. Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, United States of America
| | - Helim Aranda-Espinoza
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, United States of America
- * E-mail:
| |
Collapse
|
21
|
Structure and biomechanics of the endothelial transcellular circumferential invasion array in tumor invasion. PLoS One 2014; 9:e89758. [PMID: 24587014 PMCID: PMC3933692 DOI: 10.1371/journal.pone.0089758] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 01/24/2014] [Indexed: 11/19/2022] Open
Abstract
Cancer cells breach the endothelium not only through cell-cell junctions but also via individual endothelial cells (ECs), or transcellular invasion. The underlying EC forms a circular structure around the transcellular invasion pore that is dependent on myosin light chain kinase (MLCK) and myosin II regulatory light chain (RLC) phosphorylation. Here we offer mechanistic insights into transcellular invasive array formation amid persistent tensile force from activated EC myosin. Fluorescence recovery after photobleaching (FRAP) experiments, sarcomeric distance measurements using super-resolution microscopy and electron microscopy provide details about the nature of the myosin II invasion array. To probe the relationship between biomechanical forces and the tension required to maintain the curvature of contractile filaments, we targeted individual actin-myosin fibers at the invasion site for photoablation. We showed that adjacent filaments rapidly replace the ablat11ed structures. We propose that the transcellular circumferential invasion array (TCIA) provides the necessary constraint within the EC to blunt the radial compression from the invading cancer cell.
Collapse
|
22
|
Raman PS, Paul CD, Stroka KM, Konstantopoulos K. Probing cell traction forces in confined microenvironments. LAB ON A CHIP 2013; 13:4599-607. [PMID: 24100608 PMCID: PMC5409513 DOI: 10.1039/c3lc50802a] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Cells migrate in vivo within three-dimensional (3D) extracellular matrices. Cells also migrate through 3D longitudinal channels formed between the connective tissue and the basement membrane of muscle, nerve, and epithelium. Although traction forces have been measured during 2D cell migration, no assay has been developed to probe forces during migration through confined microenvironments. We thus fabricated a novel microfluidic device consisting of deflectable PDMS microposts incorporated within microchannels of varying cross-sectional areas. Using NIH-3T3 fibroblasts and human osteosarcoma (HOS) cells as models, we found that the average traction forces per post decreased upon increasing confinement. Inhibition of myosin-II function by blebbistatin in HOS cells decreased traction forces in unconfined (wide) channels but failed to alter them in confined spaces. Myosin activation by calyculin A also failed to affect traction forces in confining channels but increased them in wide channels. These observations underlie the importance of the physical microenvironment in the regulation of cell migration and cellular traction forces.
Collapse
Affiliation(s)
- Phrabha S. Raman
- Johns Hopkins Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA
| | - Colin D. Paul
- Johns Hopkins Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA
- Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA
| | - Kimberly M. Stroka
- Johns Hopkins Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA
- Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA
| | - Konstantinos Konstantopoulos
- Johns Hopkins Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA
- Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA
- Center of Cancer Nanotechnology Excellence, The Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA
- ; Fax: +1 410 516 5510; Tel: +1 410 516 7170
| |
Collapse
|
23
|
Lee PF, Bai Y, Smith RL, Bayless KJ, Yeh AT. Angiogenic responses are enhanced in mechanically and microscopically characterized, microbial transglutaminase crosslinked collagen matrices with increased stiffness. Acta Biomater 2013; 9:7178-90. [PMID: 23571003 DOI: 10.1016/j.actbio.2013.04.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 03/12/2013] [Accepted: 04/01/2013] [Indexed: 01/25/2023]
Abstract
During angiogenesis, endothelial cells (ECs) use both soluble and insoluble cues to expand the existing vascular network to meet the changing trophic needs of the tissue. Fundamental to this expansion are physical interactions between ECs and extracellular matrix (ECM) that influence sprout migration, lumen formation and stabilization. These physical interactions suggest that ECM mechanical properties may influence sprouting ECs and, therefore, angiogenic responses. In a three-dimensional angiogenic model in which a monolayer of ECs is induced to invade an underlying collagen matrix, angiogenic responses were measured as a function of collagen matrix stiffness by inducing collagen crosslinking with microbial transglutaminase (mTG). By biaxial mechanical testing, stiffer collagen matrices were measured with both mTG treatment and incubation time. Using two-photon excited fluorescence (TPF) and second harmonic generation (SHG), it was shown that collagen TPF intensity increased with mTG treatment, and the TPF/SHG ratio correlated with biaxially tested mechanical stiffness. SHG and OCM were further used to show that other ECM physical properties such as porosity and pore size did not change with mTG treatment, thus verifying that matrix stiffness was tuned independently of matrix density. The results showed that stiffer matrices promote more angiogenic sprouts that invade deeper. No differences in lumen size were observed between control and mTG stiffened matrices, but greater remodeling was revealed in stiffer gels using SHG and OCM. The results of this study show that angiogenic responses are influenced by stiffness and suggest that ECM properties may be useful in regenerative medicine applications to engineer angiogenesis.
Collapse
Affiliation(s)
- P-F Lee
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | | | | | | | | |
Collapse
|
24
|
Human neutrophil cytoskeletal dynamics and contractility actively contribute to trans-endothelial migration. PLoS One 2013; 8:e61377. [PMID: 23626676 PMCID: PMC3634075 DOI: 10.1371/journal.pone.0061377] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 03/08/2013] [Indexed: 01/13/2023] Open
Abstract
Transmigration through the endothelium is a key step in the immune response. In our recent work, the mechanical properties of the subendothelial matrix and biophysical state of the endothelium have been identified as key modulators of leukocyte trans-endothelial migration. Here, we demonstrated that neutrophil contractile forces and cytoskeletal dynamics also play an active biophysical role during transmigration through endothelial cell-cell junctions. Using our previously-established model for leukocyte transmigration, we first discovered that >93% of human neutrophils preferentially exploit the paracellular mode of transmigration in our in vitro model, and that is independent of subendothelial matrix stiffness. We demonstrated that inhibition of actin polymerization or depolymerization completely blocks transmigration, thus establishing a critical role for neutrophil actin dynamics in transmigration. Next, inhibition of neutrophil myosin II-mediated contractile forces renders 44% of neutrophils incapable of retracting their trailing edge under the endothelium for several minutes after the majority of the neutrophil transmigrates. Meanwhile, inhibition of neutrophil contractile forces or stabilization of microtubules doubles the time to complete transmigration for the first neutrophils to cross the endothelium. Notably, the time to complete transmigration is significantly reduced for subsequent neutrophils that cross through the same path as a previous neutrophil and is less dependent on neutrophil contractile forces and microtubule dynamics. These results suggest that the first neutrophil induces a gap in endothelial cell-cell adhesions, which “opens the door” in the endothelium and facilitates transmigration of subsequent neutrophils through the same hole. Collectively, this work demonstrates that neutrophils play an active biophysical role during the transmigration step of the immune response.
Collapse
|
25
|
Xu T, Yue W, Li CW, Yao X, Yang M. Microfluidics study of intracellular calcium response to mechanical stimulation on single suspension cells. LAB ON A CHIP 2013; 13:1060-9. [PMID: 23403699 DOI: 10.1039/c3lc40880a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
A microfluidic microdevice was developed to exert mechanical stimulation on an individual suspension cell for mechanosensation research. In this microfluidic chip, an individual cell was isolated from a population of cells, and trapped in a microchannel with a compressive component made of a deflectable membrane. The mechanosensation of HL60 cells (leukemic cells) was studied using this chip, and the results showed that mechanical stimulations could trigger extracellular calcium to flow into HL60 cells through ion channels on cell membranes. The tension on individual HL60 cells exerted by the microdevice was showed large variations in the threshold of mechanosensation activation. In contrast to previous reports using patch clamp technique, there was little influence of cytoskeleton interruption on HL60 cell mechanosensation triggered by whole-cell compression. Additionally, two functional units were integrated in one chip for carrying out mechanosensation study in parallel, where HL60 cells (leukemic cells) and Jurkat cells (lymphocytes) were shown to respond to mechanical stimulation with different kinetics. The results demonstrated that the microfluidic device provides a novel approach to investigating the mechanosensation of single suspension cells in high-throughput.
Collapse
Affiliation(s)
- Tao Xu
- Department of Biology and Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, China
| | | | | | | | | |
Collapse
|
26
|
Bianchi E, Molteni R, Pardi R, Dubini G. Microfluidics for in vitro biomimetic shear stress-dependent leukocyte adhesion assays. J Biomech 2012. [PMID: 23200903 DOI: 10.1016/j.jbiomech.2012.10.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Recruitment of leukocytes from blood to tissues is a multi-step process playing a major role in the activation of inflammatory responses. Tethering and rolling of leukocytes along the vessel wall, followed by arrest and transmigration through the endothelium result from chemoattractant-dependent signals, inducing adhesive and migratory events. Shear forces exerted by the blood flow on leukocytes induce rolling via selectin-mediated interactions with endothelial cells and increase the probability of leukocytes to engage their chemokine receptors, facilitating integrin activation and consequent arrest. Flow-derived shear forces generate mechanical stimuli concurring with biochemical signals in the modulation of leukocyte-endothelial cell interactions. In the last few years, a host of in vitro studies have clarified the biochemical adhesion cascade and the role of shear stress in leukocyte extravasation. The limitation of the static environment in Boyden devices has been overcome both by the use of parallel-plate flow chambers and by custom models mimicking the in vivo conditions, along with widespread microfluidic approaches to in vitro modeling. These devices create an in vitro biomimetic environment where the multi-step transmigration process can be imaged and quantified under mechanical and biochemical controlled conditions, including fluid dynamic settings, channel design, materials and surface coatings. This paper reviews the technological solutions recently proposed to model, observe and quantify leukocyte adhesion behavior under shear flow, with a final survey of high-throughput solutions featuring multiple parallel assays as well as thorough and time-saving statistical interpretation of the experimental results.
Collapse
Affiliation(s)
- Elena Bianchi
- LaBS-Laboratory of Biological Structure Mechanics, Department of Structural Engineering, Politecnico di Milano, Milan, Italy.
| | | | | | | |
Collapse
|
27
|
Voyvodic PL, Min D, Baker AB. A multichannel dampened flow system for studies on shear stress-mediated mechanotransduction. LAB ON A CHIP 2012; 12:3322-30. [PMID: 22836694 PMCID: PMC3426609 DOI: 10.1039/c2lc40526a] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Shear stresses are powerful regulators of cellular function and potent mediators of the development of vascular disease. We have designed and optimized a system allowing the application of flow to cultured cells in a multichannel format. By using a multichannel peristaltic pump, flow can be driven continuously in the system for long-term studies in multiple isolated flow loops. A key component of the system is a dual-chamber pulse dampener that removes the pulsatility of the flow without the need for having an open system or elevated reservoir. We optimized the design parameters of the pulse dampening chambers for the maximum reduction in flow pulsation while minimizing the fluid needed for each isolated flow channel. Human umbilical vein endothelial cells (HUVECs) were exposed to steady and pulsatile shear stress using the system. We found that cells under steady flow had a marked increased production of eNOS and formation of actin stress fibers in comparison to those under pulsatile flow conditions. Overall, the results confirm the utility of the device as a practical means to apply shear stress to cultured cells in the multichannel format and provide steady, long term flow to microfluidic devices.
Collapse
Affiliation(s)
- Peter L. Voyvodic
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX
| | - Daniel Min
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX
| | - Aaron B. Baker
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX
| |
Collapse
|
28
|
Stroka KM, Vaitkus JA, Aranda-Espinoza H. Endothelial cells undergo morphological, biomechanical, and dynamic changes in response to tumor necrosis factor-α. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2012; 41:939-47. [PMID: 22940754 DOI: 10.1007/s00249-012-0851-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 08/09/2012] [Accepted: 08/17/2012] [Indexed: 01/13/2023]
Abstract
The immune response triggers a complicated sequence of events, one of which is release of the cytokine tumor necrosis factor-α (TNF-α) from stromal cells, for example monocytes and macrophages. In this work we investigated the biophysical effects of TNF-α on endothelial cells (ECs), including changes in cell morphology, biomechanics, migration, and cytoskeletal dynamics. We found that TNF-α induces a wide distribution of cell area and aspect ratio, with these properties increasing on average during treatment. Interestingly, aspect ratio peaks after approximately 10 h of exposure to TNF-α, corresponding also to a peak in exerted traction forces. Meanwhile, ECs treated with TNF-α soften, and we associate this with significant increases in estimated cellular volume. In addition, our evaluation of migratory dynamics revealed an inverse correlation between cell aspect ratio and migration speed after TNF-α treatment, suggesting that cell shape may be an important functional regulator of EC migration during an inflammatory response. Finally, we addressed the basic mechanics of how the reorganization of F-actin filaments occurs during TNF-α treatment, and observed a dynamic shift of existing actin filaments. Together, our results suggest a functional link between EC morphology, biomechanics, migration, and cytoskeletal dynamics during an inflammatory response.
Collapse
Affiliation(s)
- Kimberly M Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA.
| | | | | |
Collapse
|
29
|
Obi S, Masuda H, Shizuno T, Sato A, Yamamoto K, Ando J, Abe Y, Asahara T. Fluid shear stress induces differentiation of circulating phenotype endothelial progenitor cells. Am J Physiol Cell Physiol 2012; 303:C595-606. [PMID: 22744008 DOI: 10.1152/ajpcell.00133.2012] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Endothelial progenitor cells (EPCs) are mobilized from bone marrow to peripheral blood, and contribute to angiogenesis in tissue. In the process, EPCs are exposed to shear stress generated by blood flow and tissue fluid flow. Our previous study showed that shear stress induces differentiation of mature EPCs in adhesive phenotype into mature endothelial cells and, moreover, arterial endothelial cells. In this study we investigated whether immature EPCs in a circulating phenotype differentiate into mature EPCs in response to shear stress. When floating-circulating phenotype EPCs derived from ex vivo expanded human cord blood were exposed to controlled levels of shear stress in a flow-loading device, the bioactivities of adhesion, migration, proliferation, antiapoptosis, tube formation, and differentiated type of EPC colony formation increased. The surface protein expression rate of the endothelial markers VEGF receptor 1 (VEGF-R1) and -2 (VEGF-R2), VE-cadherin, Tie2, VCAM1, integrin α(v)/β(3), and E-selectin increased in shear-stressed EPCs. The VEGF-R1, VEGF-R2, VE-cadherin, and Tie2 protein increases were dependent on the magnitude of shear stress. The mRNA levels of VEGF-R1, VEGF-R2, VE-cadherin, Tie2, endothelial nitric oxide synthase, matrix metalloproteinase 9, and VEGF increased in shear-stressed EPCs. Inhibitor analysis showed that the phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signal transduction pathway is a potent activator of adhesion, proliferation, tube formation, and differentiation in response to shear stress. Western blot analysis revealed that shear stress activated the VEGF-R2 phosphorylation in a ligand-independent manner. These results indicate that shear stress increases differentiation, adhesion, migration, proliferation, antiapoptosis, and vasculogenesis of circulating phenotype EPCs by activation of VEGF-R2 and the PI3K/Akt/mTOR signal transduction pathway.
Collapse
Affiliation(s)
- Syotaro Obi
- Department of Regenerative Medicine Science, Tokai University School of Medicine, Isehara, Japan
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Stroka KM, Levitan I, Aranda-Espinoza H. OxLDL and substrate stiffness promote neutrophil transmigration by enhanced endothelial cell contractility and ICAM-1. J Biomech 2012; 45:1828-34. [PMID: 22560286 DOI: 10.1016/j.jbiomech.2012.04.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 03/05/2012] [Accepted: 04/09/2012] [Indexed: 01/28/2023]
Abstract
Elevated levels of oxLDL in the bloodstream and increased vasculature stiffness are both associated with cardiovascular disease in patients. However, it is not known how oxLDL and subendothelial matrix stiffness together regulate an immune response. Here, we used an in vitro model of the vascular endothelium to explore the combined effects of oxLDL and subendothelial matrix stiffening on neutrophil transmigration. We prepared fibronectin-coated polyacrylamide gels of varying stiffness and plated human umbilical vein endothelial cells (ECs) onto the gels. We observed that oxLDL treatment of the endothelium promoted neutrophil transmigration (from <1% to 26% on soft 0.87kPa substrates), with stiffer substrates further promoting transmigration (54% on 5kPa and 41% on 280kPa). OxLDL exposure enhanced intercellular adhesion molecule-1 (ICAM-1) expression on the endothelium, which was likely responsible for the oxLDL-induced transmigration. Importantly, inhibition of MLCK-mediated EC contraction reduced transmigration to ∼9% on all substrates and eliminated the effects of subendothelial matrix stiffness. In addition, large holes, thousands of square microns in size, formed in monolayers on stiff substrates following transmigration, indicating that oxLDL treatment and subsequent neutrophil transmigration caused serious damage to the endothelium. Our results reveal that an interplay between ICAM-1 and MLCK-dependent contractile forces mediates neutrophil transmigration through oxLDL-treated endothelium. Thus, microvasculature stiffness, which likely varies depending on tissue location and health, is an important regulator of the transmigration step of the immune response in the presence of oxLDL.
Collapse
Affiliation(s)
- Kimberly M Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | | | | |
Collapse
|
31
|
Porat Z, Yaron I, Katz BZ, Kam Z, Geiger B. Shear flow-induced formation of tubular cell protrusions in multiple myeloma cells. J Cell Physiol 2011; 226:3197-207. [PMID: 21344380 DOI: 10.1002/jcp.22680] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Exposure of live cells to shear flow induces major changes in cell shape, adhesion to the extracellular matrix, and migration. In the present study, we show that exposure of cultured multiple myeloma (MM) cells to shear flow of 4-36 dynes/cm(2) triggers the extension of long tubular protrusions (denoted flow-induced protrusions, or FLIPs) in the direction of the flow. These FLIPs were found to be rich in actin, contain few or no microtubules and, apart from endoplasmic reticulum (ER)-like membranal structures, are devoid of organelles. Studying the dynamics of this process revealed that FLIPs elongate at their tips in a shear force-dependent manner, and retract at their bases. Examination of this force dependence revealed considerable heterogeneity in the mechanosensitivity of individual cells, most likely reflecting the diversity of the malignant B cell population. The mechanisms underlying FLIP formation following mechanical perturbation, and their relevance to the cellular trafficking of MM cells, are discussed.
Collapse
Affiliation(s)
- Ziv Porat
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | | | | | | | | |
Collapse
|
32
|
Mihai C, Bao S, Lai JP, Ghadiali SN, Knoell DL. PTEN inhibition improves wound healing in lung epithelia through changes in cellular mechanics that enhance migration. Am J Physiol Lung Cell Mol Physiol 2011; 302:L287-99. [PMID: 22037358 DOI: 10.1152/ajplung.00037.2011] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The phosphoinositide-3 kinase/Akt pathway is a vital survival axis in lung epithelia. We previously reported that inhibition of phosphatase and tensin homolog deleted on chromosome 10 (PTEN), a major suppressor of this pathway, results in enhanced wound repair following injury. However, the precise cellular and biomechanical mechanisms responsible for increased wound repair during PTEN inhibition are not yet well established. Using primary human lung epithelia and a related lung epithelial cell line, we first determined whether changes in migration or proliferation account for wound closure. Strikingly, we observed that cell migration accounts for the majority of wound recovery following PTEN inhibition in conjunction with activation of the Akt and ERK signaling pathways. We then used fluorescence and atomic force microscopy to investigate how PTEN inhibition alters the cytoskeletal and mechanical properties of the epithelial cell. PTEN inhibition did not significantly alter cytoskeletal structure but did result in large spatial variations in cell stiffness and in particular a decrease in cell stiffness near the wound edge. Biomechanical changes, as well as migration rates, were mediated by both the Akt and ERK pathways. Our results indicate that PTEN inhibition rapidly alters biochemical signaling events that in turn provoke alterations in biomechanical properties that enhance cell migration. Specifically, the reduced stiffness of PTEN-inhibited cells promotes larger deformations, resulting in a more migratory phenotype. We therefore conclude that increased wound closure consequent to PTEN inhibition occurs through enhancement of cell migration that is due to specific changes in the biomechanical properties of the cell.
Collapse
Affiliation(s)
- Cosmin Mihai
- Department of Biomedical Engineering, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, The Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | | | |
Collapse
|
33
|
Zeng Y, Shen Y, Huang XL, Liu XJ, Liu XH. Roles of mechanical force and CXCR1/CXCR2 in shear-stress-induced endothelial cell migration. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2011; 41:13-25. [PMID: 21989491 DOI: 10.1007/s00249-011-0752-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 09/13/2011] [Indexed: 02/05/2023]
Abstract
We previously demonstrated that CXCR1 and CXCR2 are novel mechanosensors mediating laminar shear-stress-induced endothelial cell (EC) migration (Zeng et al. in Cytokine 53:42-51, 2011). In the present study, an analytical model was proposed to further analyze the underlying mechanisms, assuming the mechanical force (MF) and mechanosensor-mediated biochemical reactions induce cell migration together. Shear stress can regulate both mechanosensor-mediated migration in the flow direction (Ms-M(FD)) and mechanosensor-mediated migration toward a wound (Ms-M(W)). Next, the migration distance, the roles of MF-induced cell migration (MF-M), and the mobilization mechanisms of mechanosensors were analyzed. The results demonstrated that MF-M plays an important role in 15.27 dyn/cm(2) shear-stress-induced EC migration but is far weaker than Ms-M(W) at 5.56 dyn/cm(2). Our findings also indicated that CXCR2 played a primary role, in synergy with CXCR1. The Ms-M(FD) was primarily mediated by the synergistic effect of CXCR1 and CXCR2. In Ms-M(W), when shear stress was beyond a certain threshold, the synergistic effect of CXCR1 and CXCR2 was enhanced, and the effect of CXCR1 was inhibited. Therefore, the retarding of EC migration and wound closure capacity under low shear flow was related to the low magnitude of shear stress, which may contribute to atherogenesis and many other vascular diseases.
Collapse
Affiliation(s)
- Ye Zeng
- Laboratory of Cardiovascular Diseases, West China Hospital, Sichuan University, No.17 Renmin Nanlu 3 Duan, Chengdu, People's Republic of China
| | | | | | | | | |
Collapse
|
34
|
Stroka KM, Aranda-Espinoza H. Endothelial cell substrate stiffness influences neutrophil transmigration via myosin light chain kinase-dependent cell contraction. Blood 2011; 118:1632-40. [PMID: 21652678 PMCID: PMC3156049 DOI: 10.1182/blood-2010-11-321125] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Accepted: 05/19/2011] [Indexed: 11/20/2022] Open
Abstract
A vast amount of work has been dedicated to the effects of shear flow and cytokines on leukocyte transmigration. However, no studies have explored the effects of substrate stiffness on transmigration. Here, we investigated important aspects of endothelial cell contraction-mediated neutrophil transmigration using an in vitro model of the vascular endothelium. We modeled blood vessels of varying mechanical properties using fibronectin-coated polyacrylamide gels of varying physiologic stiffness, plated with human umbilical vein endothelial cell (HUVEC) monolayers, which were activated with tumor necrosis factor-α. Interestingly, neutrophil transmigration increased with increasing substrate stiffness below the endothelium. HUVEC intercellular adhesion molecule-1 expression, stiffness, cytoskeletal arrangement, morphology, and cell-substrate adhesion could not account for the dependence of transmigration on HUVEC substrate stiffness. We also explored the role of cell contraction and observed that large holes formed in endothelium on stiff substrates several minutes after neutrophil transmigration reached a maximum. Further, suppression of contraction through inhibition of myosin light chain kinase normalized the effects of substrate stiffness by reducing transmigration and eliminating hole formation in HUVECs on stiff substrates. These results provide strong evidence that neutrophil transmigration is regulated by myosin light chain kinase-mediated endothelial cell contraction and that this event depends on subendothelial cell matrix stiffness.
Collapse
Affiliation(s)
- Kimberly M Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD, USA
| | | |
Collapse
|
35
|
Dietrich M, Bartfeld S, Munke R, Lange C, Ogilvie LA, Friedrich A, Meyer TF. Activation of NF-κB by Neisseria gonorrhoeae is associated with microcolony formation and type IV pilus retraction. Cell Microbiol 2011; 13:1168-82. [PMID: 21615661 DOI: 10.1111/j.1462-5822.2011.01607.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The early stage of infection with Neisseria gonorrhoeae (Ngo), the causative agent of gonorrhoea, is marked by type IV pilus (Tfp)-mediated attachment and the formation of bacterial microcolonies on epithelial cells. Retraction of the Ngo Tfp generates substantial force on its substrate which can elicit host cell signalling. Here, we observed that this retraction force could also activate nuclear factor (NF)-κB, the central signalling cascade of innate immunity. Using a p65-GFP-expressing epithelial cell line, we show that piliated Ngo induce asynchronous NF-κB activation in infected cells, which is temporally associated with the formation of gonococcal microcolonies. A mutant lacking PilT, an ATPase necessary for Tfp retraction, induced markedly reduced NF-κB activation. This was accompanied by decreased NF-κB target gene transcription and cytokine release. The impaired ability of the pilT mutant to activate NF-κB was compensated by applying mechanical shear stress to the infected host cells, indicating that the mechanical forces generated by retractile pili are involved in the retraction-dependent activation of NF-κB elicited by gonococcal microcolonies. Thus, our work provides evidence for an intriguing relationship between microcolony growth, pilus retraction and host cell signalling, with likely implications with regard to the course of symptomatic versus asymptomatic gonococcal infections.
Collapse
Affiliation(s)
- Manuela Dietrich
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
36
|
Hidden malignant cells within leukocyte aggregates: seeds for invasive and metastatic cancer? Cancer Epidemiol 2011; 35:475-9. [PMID: 21292584 DOI: 10.1016/j.canep.2010.11.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2010] [Revised: 11/24/2010] [Accepted: 11/25/2010] [Indexed: 11/20/2022]
Abstract
BACKGROUND Our previous studies revealed that leukocyte infiltration into aged or injured myoepithelial cell layers is a key trigger for breast tumor invasion and metastasis. Our current study further assessed the possibility that leukocyte aggregates may harbor detached individual tumor cell or clusters of tumor cells. MATERIALS AND METHODS Tissue sections from patients with pregnancy-associated breast cancer (PABC) and controls were subjected to morphological and immunohistochemical assessment with a panel of leukocyte and tumor cell related markers. RESULTS A total of 63 leukocyte aggregates were detected in the 20 PABC cases studied. Of these, 55 (87%) were distributed within normal or hyperplastic lobules adjacent to invasive lesions. Over 70% of these leukocyte aggregates harbored detached individual tumor cell or cell clusters with malignant properties, including strong p53 positivity, elevated proliferation, reduced cell surface adhesion molecules, and cytological resemblance to adjacent invasive cancer cells. A significant number of these tumor cells or condensed chromosomes of mitotic tumor cells were observed to conjoin with the plasma membrane of leukocytes. Similar alterations were seen in leukocyte aggregates within the inter-lobular space and in non-PABC with a lower frequency. CONCLUSIONS These findings suggest that leukocyte infiltration may trigger dissemination of tumor cells from their primary site, and that leukocyte aggregates may serve as a reservoir for disseminated tumor cells that may be physically dragged to distant sites by leukocytes during their migration.
Collapse
|
37
|
Stroka KM, Aranda-Espinoza H. Effects of Morphology vs. Cell-Cell Interactions on Endothelial Cell Stiffness. Cell Mol Bioeng 2010; 4:9-27. [PMID: 21359128 DOI: 10.1007/s12195-010-0142-y] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Biological processes such as atherogenesis, wound healing, cancer cell metastasis, and immune cell transmigration rely on a delicate balance between Cell-Cell and cell-substrate adhesion. Cell mechanics have been shown to depend on substrate factors such as stiffness and ligand presentation, while the effects of Cell-Cell interactions on the mechanical properties of cells has received little attention. Here, we use atomic force microscopy to measure the Young's modulus of live human umbilical vein endothelial cells (HUVECs). In varying the degree of Cell-Cell contact in HUVECs (single cells, groups, and monolayers), we observe that increased cell stiffness correlates with an increase in cell area. Further, we observe that HUVECs stiffen as they spread onto a glass substrate. When we weaken Cell-Cell junctions (i.e., through a low dose of cytochalasin B or treatment with a VE-cadherin antibody), we observe that cell-substrate adhesion increases, as measured by focal adhesion size and density, and the stiffness of cells within the monolayer approaches that of single cells. Our results suggest that while morphology can roughly be used to predict cell stiffness, Cell-Cell interactions may play a significant role in determining the mechanical properties of individual cells in tissues by careful maintenance of cell tension homeostasis.
Collapse
Affiliation(s)
- Kimberly M Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, 3138 Jeong H. Kim Engineering Building, College Park, MD 20742, USA
| | | |
Collapse
|