1
|
Nam K, Torkzaban M, Shames JP, Liao L, Wessner CE, Machado P, Lyshchik A, Forsberg F. Characterization of Indeterminate Breast Lesions Based on Pressure Estimates by Noninvasive 3D Contrast-Enhanced Ultrasound. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:1854-1860. [PMID: 39237425 PMCID: PMC11490378 DOI: 10.1016/j.ultrasmedbio.2024.08.010] [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: 02/28/2024] [Revised: 07/15/2024] [Accepted: 08/10/2024] [Indexed: 09/07/2024]
Abstract
OBJECTIVE To assess the ability of the pressure gradient between breast lesions and adjacent normal tissue estimated by 3D subharmonic-aided pressure estimation (SHAPE) to characterize indeterminate breast lesions. METHODS This prospective study enrolled patients scheduled for ultrasound-guided needle biopsies of a breast lesion. Before the biopsy, 3D SHAPE data were collected from the breast lesion during the infusion of an ultrasound contrast agent (Definity) as well as after clearance of the agent. Direct, invasive pressure measurements in the lesion and adjacent normal tissue were then obtained using an intracompartmental pressure monitoring system (C2DX) before tissue sampling as part of the biopsy procedure. The mean SHAPE gradient and invasive measurement gradient between the lesion and adjacent normal tissue were compared to the biopsy results. The SHAPE gradients were also compared to the invasive pressure gradients. RESULTS There were 8 malignant and 13 benign lesions studied. The SHAPE gradients and invasive pressure gradients were significantly different between the benign and malignant lesions (2.86 ± 3.24 vs. -0.03 ± 1.72 a.u.; p = 0.03 and 9.9 ± 8.5 vs. 20.9 ± 8.0 mmHg; p = 0.008, respectively). The area under the curves, specificities, and sensitivities for detecting malignancy by SHAPE gradients and invasive pressure gradients were 0.79 and 0.88, 77% and 92%, and 88% and 50%, respectively. A weak negative correlation was found between the SHAPE and invasive pressure gradients (r = -0.2). CONCLUSION The pressure gradient between a breast lesion and adjacent normal tissue estimated by 3D SHAPE shows potential for characterizing indeterminate breast lesions.
Collapse
Affiliation(s)
- Kibo Nam
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA.
| | - Mehnoosh Torkzaban
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jason P Shames
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Lydia Liao
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Corinne E Wessner
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Priscilla Machado
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Andrej Lyshchik
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Flemming Forsberg
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
2
|
Crouigneau R, Li YF, Auxillos J, Goncalves-Alves E, Marie R, Sandelin A, Pedersen SF. Mimicking and analyzing the tumor microenvironment. CELL REPORTS METHODS 2024; 4:100866. [PMID: 39353424 PMCID: PMC11573787 DOI: 10.1016/j.crmeth.2024.100866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 07/22/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024]
Abstract
The tumor microenvironment (TME) is increasingly appreciated to play a decisive role in cancer development and response to therapy in all solid tumors. Hypoxia, acidosis, high interstitial pressure, nutrient-poor conditions, and high cellular heterogeneity of the TME arise from interactions between cancer cells and their environment. These properties, in turn, play key roles in the aggressiveness and therapy resistance of the disease, through complex reciprocal interactions between the cancer cell genotype and phenotype, and the physicochemical and cellular environment. Understanding this complexity requires the combination of sophisticated cancer models and high-resolution analysis tools. Models must allow both control and analysis of cellular and acellular TME properties, and analyses must be able to capture the complexity at high depth and spatial resolution. Here, we review the advantages and limitations of key models and methods in order to guide further TME research and outline future challenges.
Collapse
Affiliation(s)
- Roxane Crouigneau
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Yan-Fang Li
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Jamie Auxillos
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Eliana Goncalves-Alves
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Rodolphe Marie
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Albin Sandelin
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark.
| | - Stine Falsig Pedersen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
3
|
Amiri F, Akinpelu AA, Keith WC, Hemmati F, Vaghasiya RS, Bowen D, Waliagha RS, Wang C, Chen P, Mitra AK, Li Y, Mistriotis P. Confinement controls the directional cell responses to fluid forces. Cell Rep 2024; 43:114692. [PMID: 39207902 PMCID: PMC11495937 DOI: 10.1016/j.celrep.2024.114692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/18/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
Our understanding of how fluid forces influence cell migration in confining environments remains limited. By integrating microfluidics with live-cell imaging, we demonstrate that cells in tightly-but not moderately-confined spaces reverse direction and move upstream upon exposure to fluid forces. This fluid force-induced directional change occurs less frequently when cells display diminished mechanosensitivity, experience elevated hydraulic resistance, or sense a chemical gradient. Cell reversal requires actin polymerization to the new cell front, as shown mathematically and experimentally. Actin polymerization is necessary for the fluid force-induced activation of NHE1, which cooperates with calcium to induce upstream migration. Calcium levels increase downstream, mirroring the subcellular distribution of myosin IIA, whose activation enhances upstream migration. Reduced lamin A/C levels promote downstream migration of metastatic tumor cells by preventing cell polarity establishment and intracellular calcium rise. This mechanism could allow cancer cells to evade high-pressure environments, such as the primary tumor.
Collapse
Affiliation(s)
- Farshad Amiri
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA
| | - Ayuba A Akinpelu
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA
| | - William C Keith
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA
| | - Farnaz Hemmati
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA
| | - Ravi S Vaghasiya
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA
| | - Dylan Bowen
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA
| | - Razan S Waliagha
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA
| | - Chuanyu Wang
- Materials Research and Education Center, Auburn University, Auburn, AL 36849, USA
| | - Pengyu Chen
- Materials Research and Education Center, Auburn University, Auburn, AL 36849, USA
| | - Amit K Mitra
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA; Center for Pharmacogenomics and Single-Cell Omics (AUPharmGx), Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA; UAB O'Neal Comprehensive Cancer, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35233, USA
| | - Yizeng Li
- Department of Biomedical Engineering, Binghamton University, SUNY, Binghamton, NY 13902, USA
| | | |
Collapse
|
4
|
Liu Z, Fan Y, Cui M, Wang X, Zhao P. Investigation of tumour environments through advancements in microtechnology and nanotechnology. Biomed Pharmacother 2024; 178:117230. [PMID: 39116787 DOI: 10.1016/j.biopha.2024.117230] [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: 06/05/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
Cancer has a significant negative social and economic impact on both developed and developing countries. As a result, understanding the onset and progression of cancer is critical for developing therapies that can improve the well-being and health of individuals with cancer. With time, study has revealed, the tumor microenvironment has great influence on this process. Micro and nanoscale engineering techniques can be used to study the tumor microenvironment. Nanoscale and Microscale engineering use Novel technologies and designs with small dimensions to recreate the TME. Knowing how cancer cells interact with one another can help researchers develop therapeutic approaches that anticipate and counteract cancer cells' techniques for evading detection and fighting anti-cancer treatments, such as microfabrication techniques, microfluidic devices, nanosensors, and nanodevices used to study or recreate the tumor microenvironment. Nevertheless, a complicated action just like the growth and in cancer advancement, and their intensive association along the environment around it that has to be studied in more detail.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Radiology, Shengjing Hospital of China Medical University, China
| | - Yan Fan
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Mengyao Cui
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xu Wang
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Pengfei Zhao
- Department of Radiology, Shengjing Hospital of China Medical University, China.
| |
Collapse
|
5
|
Chen C, Boché A, Wang Z, Lopez E, Peng J, Carreiras F, Schanne-Klein MC, Chen Y, Lambert A, Aimé C. The Balance Between Shear Flow and Extracellular Matrix in Ovarian Cancer-on-Chip. Adv Healthc Mater 2024; 13:e2400938. [PMID: 38829702 DOI: 10.1002/adhm.202400938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/30/2024] [Indexed: 06/05/2024]
Abstract
Ovarian cancer is the most lethal gynecologic cancer in developed countries. In the tumor microenvironment, the extracellular matrix (ECM) and flow shear stress are key players in directing ovarian cancer cells invasion. Artificial ECM models based only on ECM proteins are used to build an ovarian tumor-on-chip to decipher the crosstalk between ECM and shear stress on the migratory behavior and cellular heterogeneity of ovarian tumor cells. This work shows that in the shear stress regime of the peritoneal cavity, the ECM plays a major role in driving individual or collective ovarian tumor cells migration. In the presence of basement membrane proteins, migration is more collective than on type I collagen regardless of shear stress. With increasing shear stress, individual cell migration is enhanced; while, no significant impact on collective migration is measured. This highlights the central position that ECM and flow shear stress should hold in in vitro ovarian cancer models to deepen understanding of cellular responses and improve development of ovarian cancer therapeutic platforms. In this frame, adding flow provides significant improvement in biological relevance over the authors' previous work. Further steps for enhanced clinical relevance require not only multiple cell lines but also patient-derived cells and sera.
Collapse
Affiliation(s)
- Changchong Chen
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, Paris, 75005, France
| | - Alphonse Boché
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, ERRMECe (EA1391), Groupe Matrice Extracellulaire et physiopathologie (MECuP), Institut des Matériaux, I-MAT (FD4122), CY Cergy Paris Université, Cergy, 95000, France
| | - Zixu Wang
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, Paris, 75005, France
| | - Elliot Lopez
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, Paris, 75005, France
| | - Juan Peng
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, Paris, 75005, France
| | - Franck Carreiras
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, ERRMECe (EA1391), Groupe Matrice Extracellulaire et physiopathologie (MECuP), Institut des Matériaux, I-MAT (FD4122), CY Cergy Paris Université, Cergy, 95000, France
| | - Marie-Claire Schanne-Klein
- Laboratoire d'Optique et Biosciences (LOB), École polytechnique, CNRS, Inserm, Institut Polytechnique de Paris, Palaiseau, F-91128, France
| | - Yong Chen
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, Paris, 75005, France
| | - Ambroise Lambert
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, ERRMECe (EA1391), Groupe Matrice Extracellulaire et physiopathologie (MECuP), Institut des Matériaux, I-MAT (FD4122), CY Cergy Paris Université, Cergy, 95000, France
| | - Carole Aimé
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, Paris, 75005, France
| |
Collapse
|
6
|
Suh YJ, Li AT, Pandey M, Nordmann CS, Huang YL, Wu M. Decoding physical principles of cell migration under controlled environment using microfluidics. BIOPHYSICS REVIEWS 2024; 5:031302. [PMID: 39091432 PMCID: PMC11290890 DOI: 10.1063/5.0199161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 06/26/2024] [Indexed: 08/04/2024]
Abstract
Living cells can perform incredible tasks that man-made micro/nano-sized robots have not yet been able to accomplish. One example is that white blood cells can sense and move to the site of pathogen attack within minutes. The robustness and precision of cellular functions have been perfected through billions of years of evolution. In this context, we ask the question whether cells follow a set of physical principles to sense, adapt, and migrate. Microfluidics has emerged as an enabling technology for recreating well-defined cellular environment for cell migration studies, and its ability to follow single cell dynamics allows for the results to be amenable for theoretical modeling. In this review, we focus on the development of microfluidic platforms for recreating cellular biophysical (e.g., mechanical stress) and biochemical (e.g., nutrients and cytokines) environments for cell migration studies in 3D. We summarize the basic principles that cells (including bacteria, algal, and mammalian cells) use to respond to chemical gradients learned from microfluidic systems. We also discuss about novel biological insights gained from studies of cell migration under biophysical cues and the need for further quantitative studies of cell function under well-controlled biophysical environments in the future.
Collapse
Affiliation(s)
- Young Joon Suh
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Alan T. Li
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Mrinal Pandey
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Cassidy S. Nordmann
- Department of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Yu Ling Huang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Mingming Wu
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| |
Collapse
|
7
|
Brooks A, Zhang Y, Chen J, Zhao CX. Cancer Metastasis-on-a-Chip for Modeling Metastatic Cascade and Drug Screening. Adv Healthc Mater 2024; 13:e2302436. [PMID: 38224141 DOI: 10.1002/adhm.202302436] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 01/06/2024] [Indexed: 01/16/2024]
Abstract
Microfluidic chips are valuable tools for studying intricate cellular and cell-microenvironment interactions. Traditional in vitro cancer models lack accuracy in mimicking the complexities of in vivo tumor microenvironment. However, cancer-metastasis-on-a-chip (CMoC) models combine the advantages of 3D cultures and microfluidic technology, serving as powerful platforms for exploring cancer mechanisms and facilitating drug screening. These chips are able to compartmentalize the metastatic cascade, deepening the understanding of its underlying mechanisms. This article provides an overview of current CMoC models, focusing on distinctive models that simulate invasion, intravasation, circulation, extravasation, and colonization, and their applications in drug screening. Furthermore, challenges faced by CMoC and microfluidic technologies are discussed, while exploring promising future directions in cancer research. The ongoing development and integration of these models into cancer studies are expected to drive transformative advancements in the field.
Collapse
Affiliation(s)
- Anastasia Brooks
- School of Chemical Engineering, University of Adelaide, Adelaide, 5005, Australia
| | - Yali Zhang
- School of Chemical Engineering, University of Adelaide, Adelaide, 5005, Australia
| | - Jiezhong Chen
- School of Chemical Engineering, University of Adelaide, Adelaide, 5005, Australia
| | - Chun-Xia Zhao
- School of Chemical Engineering, University of Adelaide, Adelaide, 5005, Australia
| |
Collapse
|
8
|
Doherty EL, Krohn G, Warren EC, Patton A, Whitworth CP, Rathod M, Biehl A, Aw WY, Freytes DO, Polacheck WJ. Human Cell-Derived Matrix Composite Hydrogels with Diverse Composition for Use in Vasculature-on-chip Models. Adv Healthc Mater 2024; 13:e2400192. [PMID: 38518808 PMCID: PMC11281875 DOI: 10.1002/adhm.202400192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/19/2024] [Indexed: 03/24/2024]
Abstract
Microphysiological and organ-on-chip platforms seek to address critical gaps in human disease models and drug development that underlie poor rates of clinical success for novel interventions. While the fabrication technology and model cells used to synthesize organs-on-chip have advanced considerably, most platforms rely on animal-derived or synthetic extracellular matrix as a cell substrate, limiting mimicry of human physiology and precluding use in modeling diseases in which matrix dynamics play a role in pathogenesis. Here, the development of human cell-derived matrix (hCDM) composite hydrogels for use in 3D microphysiologic models of the vasculature is reported. hCDM composite hydrogels are derived from human donor fibroblasts and maintain a complex milieu of basement membrane, proteoglycans, and nonfibrillar matrix components. The use of hCDM composite hydrogels as 2D and 3D cell culture substrates is demonstrated, and hCDM composite hydrogels are patterned to form engineered human microvessels. Interestingly, hCDM composite hydrogels are enriched in proteins associated with vascular morphogenesis as determined by mass spectrometry, and functional analysis demonstrates proangiogenic signatures in human endothelial cells cultured in these hydrogels. In conclusion, this study suggests that human donor-derived hCDM composite hydrogels could address technical gaps in human organs-on-chip development and serve as substrates to promote vascularization.
Collapse
Affiliation(s)
- Elizabeth L Doherty
- The Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, 10010 Mary Ellen Jones Building, 116 Manning Drive, Chapel Hill, NC 27514, USA
| | - Grace Krohn
- The Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, 10010 Mary Ellen Jones Building, 116 Manning Drive, Chapel Hill, NC 27514, USA
| | - Emily C Warren
- The Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, 10010 Mary Ellen Jones Building, 116 Manning Drive, Chapel Hill, NC 27514, USA
| | - Alexandra Patton
- The Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, 10010 Mary Ellen Jones Building, 116 Manning Drive, Chapel Hill, NC 27514, USA
| | - Chloe P Whitworth
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill School of Medicine, 130 Mason Farm Road, Chapel Hill, Carolina, NC 27599, USA
| | - Mitesh Rathod
- The Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, 10010 Mary Ellen Jones Building, 116 Manning Drive, Chapel Hill, NC 27514, USA
| | - Andreea Biehl
- The Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, 10010 Mary Ellen Jones Building, 116 Manning Drive, Chapel Hill, NC 27514, USA
| | - Wen Yih Aw
- The Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, 10010 Mary Ellen Jones Building, 116 Manning Drive, Chapel Hill, NC 27514, USA
| | - Donald O Freytes
- The Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, 10010 Mary Ellen Jones Building, 116 Manning Drive, Chapel Hill, NC 27514, USA
| | - William J Polacheck
- The Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, 10010 Mary Ellen Jones Building, 116 Manning Drive, Chapel Hill, NC 27514, USA
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, 111 Mason Farm Road, Chapel Hill, Carolina, NC 27599, USA
| |
Collapse
|
9
|
Akinpelu A, Akinsipe T, Avila LA, Arnold RD, Mistriotis P. The impact of tumor microenvironment: unraveling the role of physical cues in breast cancer progression. Cancer Metastasis Rev 2024; 43:823-844. [PMID: 38238542 PMCID: PMC11156564 DOI: 10.1007/s10555-024-10166-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 01/02/2024] [Indexed: 01/30/2024]
Abstract
Metastasis accounts for the vast majority of breast cancer-related fatalities. Although the contribution of genetic and epigenetic modifications to breast cancer progression has been widely acknowledged, emerging evidence underscores the pivotal role of physical stimuli in driving breast cancer metastasis. In this review, we summarize the changes in the mechanics of the breast cancer microenvironment and describe the various forces that impact migrating and circulating tumor cells throughout the metastatic process. We also discuss the mechanosensing and mechanotransducing molecules responsible for promoting the malignant phenotype in breast cancer cells. Gaining a comprehensive understanding of the mechanobiology of breast cancer carries substantial potential to propel progress in prognosis, diagnosis, and patient treatment.
Collapse
Affiliation(s)
- Ayuba Akinpelu
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, Auburn, AL, 36849, USA
| | - Tosin Akinsipe
- Department of Biological Sciences, College of Science and Mathematics, Auburn University, Auburn, AL, 36849, USA
| | - L Adriana Avila
- Department of Biological Sciences, College of Science and Mathematics, Auburn University, Auburn, AL, 36849, USA
| | - Robert D Arnold
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL, 36849, USA
| | - Panagiotis Mistriotis
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, Auburn, AL, 36849, USA.
| |
Collapse
|
10
|
Jackson CE, Green NH, English WR, Claeyssens F. The use of microphysiological systems to model metastatic cancer. Biofabrication 2024; 16:032002. [PMID: 38579739 DOI: 10.1088/1758-5090/ad3b70] [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: 11/13/2023] [Accepted: 04/05/2024] [Indexed: 04/07/2024]
Abstract
Cancer is one of the leading causes of death in the 21st century, with metastasis of cancer attributing to 90% of cancer-related deaths. Therefore, to improve patient outcomes there is a need for better preclinical models to increase the success of translating oncological therapies into the clinic. Current traditional staticin vitromodels lack a perfusable network which is critical to overcome the diffusional mass transfer limit to provide a mechanism for the exchange of essential nutrients and waste removal, and increase their physiological relevance. Furthermore, these models typically lack cellular heterogeneity and key components of the immune system and tumour microenvironment. This review explores rapidly developing strategies utilising perfusable microphysiological systems (MPS) for investigating cancer cell metastasis. In this review we initially outline the mechanisms of cancer metastasis, highlighting key steps and identifying the current gaps in our understanding of the metastatic cascade, exploring MPS focused on investigating the individual steps of the metastatic cascade before detailing the latest MPS which can investigate multiple components of the cascade. This review then focuses on the factors which can affect the performance of an MPS designed for cancer applications with a final discussion summarising the challenges and future directions for the use of MPS for cancer models.
Collapse
Affiliation(s)
- Caitlin E Jackson
- Materials Science and Engineering, The Kroto Research Institute, University of Sheffield, Sheffield S3 7HQ, United Kingdom
- Insigneo Institute for In Silico Medicine, The Pam Liversidge Building, University of Sheffield, Sheffield S1 3JD, United Kingdom
| | - Nicola H Green
- Materials Science and Engineering, The Kroto Research Institute, University of Sheffield, Sheffield S3 7HQ, United Kingdom
- Insigneo Institute for In Silico Medicine, The Pam Liversidge Building, University of Sheffield, Sheffield S1 3JD, United Kingdom
| | - William R English
- Norwich Medical School, University of East Anglia, Norwich NR3 7TJ, United Kingdom
| | - Frederik Claeyssens
- Materials Science and Engineering, The Kroto Research Institute, University of Sheffield, Sheffield S3 7HQ, United Kingdom
- Insigneo Institute for In Silico Medicine, The Pam Liversidge Building, University of Sheffield, Sheffield S1 3JD, United Kingdom
| |
Collapse
|
11
|
Ben-Ami Y, Pitt-Francis JM, Maini PK, Byrne HM. Using a probabilistic approach to derive a two-phase model of flow-induced cell migration. Biophys J 2024; 123:799-813. [PMID: 38414238 PMCID: PMC10995429 DOI: 10.1016/j.bpj.2024.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 01/22/2024] [Accepted: 02/21/2024] [Indexed: 02/29/2024] Open
Abstract
Interstitial fluid flow is a feature of many solid tumors. In vitro experiments have shown that such fluid flow can direct tumor cell movement upstream or downstream depending on the balance between the competing mechanisms of tensotaxis (cell migration up stress gradients) and autologous chemotaxis (downstream cell movement in response to flow-induced gradients of self-secreted chemoattractants). In this work we develop a probabilistic-continuum, two-phase model for cell migration in response to interstitial flow. We use a kinetic description for the cell velocity probability density function, and model the flow-dependent mechanical and chemical stimuli as forcing terms that bias cell migration upstream and downstream. Using velocity-space averaging, we reformulate the model as a system of continuum equations for the spatiotemporal evolution of the cell volume fraction and flux in response to forcing terms that depend on the local direction and magnitude of the mechanochemical cues. We specialize our model to describe a one-dimensional cell layer subject to fluid flow. Using a combination of numerical simulations and asymptotic analysis, we delineate the parameter regime where transitions from downstream to upstream cell migration occur. As has been observed experimentally, the model predicts downstream-oriented chemotactic migration at low cell volume fractions, and upstream-oriented tensotactic migration at larger volume fractions. We show that the locus of the critical volume fraction, at which the system transitions from downstream to upstream migration, is dominated by the ratio of the rate of chemokine secretion and advection. Our model also predicts that, because the tensotactic stimulus depends strongly on the cell volume fraction, upstream, tensotaxis-dominated migration occurs only transiently when the cells are initially seeded, and transitions to downstream, chemotaxis-dominated migration occur at later times due to the dispersive effect of cell diffusion.
Collapse
Affiliation(s)
- Yaron Ben-Ami
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK.
| | | | - Philip K Maini
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK
| | - Helen M Byrne
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK; Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK
| |
Collapse
|
12
|
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: 0] [Impact Index Per Article: 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
|
13
|
Shu J, Deng H, Zhang Y, Wu F, He J. Cancer cell response to extrinsic and intrinsic mechanical cue: opportunities for tumor apoptosis strategies. Regen Biomater 2024; 11:rbae016. [PMID: 38476678 PMCID: PMC10932484 DOI: 10.1093/rb/rbae016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 03/14/2024] Open
Abstract
Increasing studies have revealed the importance of mechanical cues in tumor progression, invasiveness and drug resistance. During malignant transformation, changes manifest in either the mechanical properties of the tissue or the cellular ability to sense and respond to mechanical signals. The major focus of the review is the subtle correlation between mechanical cues and apoptosis in tumor cells from a mechanobiology perspective. To begin, we focus on the intracellular force, examining the mechanical properties of the cell interior, and outlining the role that the cytoskeleton and intracellular organelle-mediated intracellular forces play in tumor cell apoptosis. This article also elucidates the mechanisms by which extracellular forces guide tumor cell mechanosensing, ultimately triggering the activation of the mechanotransduction pathway and impacting tumor cell apoptosis. Finally, a comprehensive examination of the present status of the design and development of anti-cancer materials targeting mechanotransduction is presented, emphasizing the underlying design principles. Furthermore, the article underscores the need to address several unresolved inquiries to enhance our comprehension of cancer therapeutics that target mechanotransduction.
Collapse
Affiliation(s)
- Jun Shu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, PR China
| | - Huan Deng
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, PR China
| | - Yu Zhang
- College of Food and Biological Engineering, Chengdu University, Chengdu 610106, PR China
| | - Fang Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, PR China
| | - Jing He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, PR China
| |
Collapse
|
14
|
Mierke CT. Phenotypic Heterogeneity, Bidirectionality, Universal Cues, Plasticity, Mechanics, and the Tumor Microenvironment Drive Cancer Metastasis. Biomolecules 2024; 14:184. [PMID: 38397421 PMCID: PMC10887446 DOI: 10.3390/biom14020184] [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: 12/25/2023] [Revised: 01/19/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Tumor diseases become a huge problem when they embark on a path that advances to malignancy, such as the process of metastasis. Cancer metastasis has been thoroughly investigated from a biological perspective in the past, whereas it has still been less explored from a physical perspective. Until now, the intraluminal pathway of cancer metastasis has received the most attention, while the interaction of cancer cells with macrophages has received little attention. Apart from the biochemical characteristics, tumor treatments also rely on the tumor microenvironment, which is recognized to be immunosuppressive and, as has recently been found, mechanically stimulates cancer cells and thus alters their functions. The review article highlights the interaction of cancer cells with other cells in the vascular metastatic route and discusses the impact of this intercellular interplay on the mechanical characteristics and subsequently on the functionality of cancer cells. For instance, macrophages can guide cancer cells on their intravascular route of cancer metastasis, whereby they can help to circumvent the adverse conditions within blood or lymphatic vessels. Macrophages induce microchannel tunneling that can possibly avoid mechanical forces during extra- and intravasation and reduce the forces within the vascular lumen due to vascular flow. The review article highlights the vascular route of cancer metastasis and discusses the key players in this traditional route. Moreover, the effects of flows during the process of metastasis are presented, and the effects of the microenvironment, such as mechanical influences, are characterized. Finally, the increased knowledge of cancer metastasis opens up new perspectives for cancer treatment.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth System Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, Leipzig University, 04103 Leipzig, Germany
| |
Collapse
|
15
|
Serrano JC, Gillrie MR, Li R, Ishamuddin SH, Moeendarbary E, Kamm RD. Microfluidic-Based Reconstitution of Functional Lymphatic Microvasculature: Elucidating the Role of Lymphatics in Health and Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302903. [PMID: 38059806 PMCID: PMC10837354 DOI: 10.1002/advs.202302903] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/17/2023] [Indexed: 12/08/2023]
Abstract
The knowledge of the blood microvasculature and its functional role in health and disease has grown significantly attributable to decades of research and numerous advances in cell biology and tissue engineering; however, the lymphatics (the secondary vascular system) has not garnered similar attention, in part due to a lack of relevant in vitro models that mimic its pathophysiological functions. Here, a microfluidic-based approach is adopted to achieve precise control over the biological transport of growth factors and interstitial flow that drive the in vivo growth of lymphatic capillaries (lymphangiogenesis). The engineered on-chip lymphatics with in vivo-like morphology exhibit tissue-scale functionality with drainage rates of interstitial proteins and molecules comparable to in vivo standards. Computational and scaling analyses of the underlying transport phenomena elucidate the critical role of the three-dimensional geometry and lymphatic endothelium in recapitulating physiological drainage. Finally, the engineered on-chip lymphatics enabled studies of lymphatic-immune interactions that revealed inflammation-driven responses by the lymphatics to recruit immune cells via chemotactic signals similar to in vivo, pathological events. This on-chip lymphatics platform permits the interrogation of various lymphatic biological functions, as well as screening of lymphatic-based therapies such as interstitial absorption of protein therapeutics and lymphatic immunomodulation for cancer therapy.
Collapse
Affiliation(s)
- Jean C. Serrano
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Mark R. Gillrie
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Medicine University of CalgaryCalgaryABT2N 1N4Canada
| | - Ran Li
- Center for Systems Biology Massachusetts General Hospital Research InstituteBostonMA02114USA
| | - Sarah H. Ishamuddin
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Emad Moeendarbary
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Mechanical EngineeringUniversity College LondonTorrington PlaceLondonWC1E 7JEUK
- 199 Biotechnologies LtdGloucester RoadLondonW2 6LDUK
| | - Roger D. Kamm
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| |
Collapse
|
16
|
Rahman Z, Bordoloi AD, Rouhana H, Tavasso M, van der Zon G, Garbin V, Ten Dijke P, Boukany PE. Interstitial flow potentiates TGF-β/Smad-signaling activity in lung cancer spheroids in a 3D-microfluidic chip. LAB ON A CHIP 2024; 24:422-433. [PMID: 38087979 PMCID: PMC10826459 DOI: 10.1039/d3lc00886j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/01/2023] [Indexed: 01/31/2024]
Abstract
Within the tumor microenvironment (TME), cancer cells use mechanotransduction pathways to convert biophysical forces to biochemical signals. However, the underlying mechanisms and functional significance of these pathways remain largely unclear. The upregulation of mechanosensitive pathways from biophysical forces such as interstitial flow (IF), leads to the activation of various cytokines, including transforming growth factor-β (TGF-β). TGF-β promotes in part via a Smad-dependent signaling pathway the epithelial-mesenchymal transition (EMT) in cancer cells. The latter process is linked to increased cancer cell motility and invasion. Current research models have limited ability to investigate the combined effects of biophysical forces (such as IF) and cytokines (TGF-β) in a 3D microenvironment. We used a 3D-matrix based microfluidic platform to demonstrate the potentiating effect of IF on exogenous TGF-β induced upregulation of the Smad-signaling activity and the expression of mesenchymal marker vimentin in A549 lung cancer spheroids. To monitor this, we used stably integrated fluorescent based reporters into the A549 cancer cell genome. Our results demonstrate that IF enhances exogenous TGF-β induced Smad-signaling activity in lung cancer spheroids embedded in a matrix microenvironment. In addition, we observed an increased cell motility for A549 spheroids when exposed to IF and TGF-β. Our 3D-microfluidic model integrated with real-time imaging provides a powerful tool for investigating cancer cell signaling and motility associated with invasion characteristics in a physiologically relevant TME.
Collapse
Affiliation(s)
- Zaid Rahman
- Department of Chemical Engineering, Delft University of Technology, Delft, The Netherlands.
| | - Ankur Deep Bordoloi
- Department of Chemical Engineering, Delft University of Technology, Delft, The Netherlands.
| | - Haifa Rouhana
- Department of Chemical Engineering, Delft University of Technology, Delft, The Netherlands.
| | - Margherita Tavasso
- Department of Chemical Engineering, Delft University of Technology, Delft, The Netherlands.
| | - Gerard van der Zon
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Valeria Garbin
- Department of Chemical Engineering, Delft University of Technology, Delft, The Netherlands.
| | - Peter Ten Dijke
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Pouyan E Boukany
- Department of Chemical Engineering, Delft University of Technology, Delft, The Netherlands.
| |
Collapse
|
17
|
Tadić V, Zhang W, Brozovic A. The high-grade serous ovarian cancer metastasis and chemoresistance in 3D models. Biochim Biophys Acta Rev Cancer 2024; 1879:189052. [PMID: 38097143 DOI: 10.1016/j.bbcan.2023.189052] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 12/21/2023]
Abstract
High-grade serous ovarian cancer (HGSOC) is the most frequent and aggressive type of epithelial ovarian cancer, with high recurrence rate and chemoresistance being the main issues in its clinical management. HGSOC is specifically challenging due to the metastatic dissemination via spheroids in the ascitic fluid. The HGSOC spheroids represent the invasive and chemoresistant cellular fraction, which is impossible to investigate in conventional two-dimensional (2D) monolayer cell cultures lacking critical cell-to-cell and cell-extracellular matrix interactions. Three-dimensional (3D) HGSOC cultures, where cells aggregate and exhibit relevant interactions, offer a promising in vitro model of peritoneal metastasis and multicellular drug resistance. This review summarizes recent studies of HGSOC in 3D culture conditions and highlights the role of multicellular HGSOC spheroids and ascitic environment in HGSOC metastasis and chemoresistance.
Collapse
Affiliation(s)
- Vanja Tadić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička Str. 54, Zagreb HR-10000, Croatia
| | - Wei Zhang
- Department of Engineering Mechanics, Dalian University of Technology, Linggong Road 2, Dalian CN-116024, China
| | - Anamaria Brozovic
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička Str. 54, Zagreb HR-10000, Croatia.
| |
Collapse
|
18
|
Buffone A, Hammer DA, Kim SHJ, Anderson NR, Mochida A, Lee DH, Guin S. Not all (cells) who wander are lost: Upstream migration as a pervasive mode of amoeboid cell motility. Front Cell Dev Biol 2023; 11:1291201. [PMID: 38020916 PMCID: PMC10651737 DOI: 10.3389/fcell.2023.1291201] [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: 09/08/2023] [Accepted: 10/06/2023] [Indexed: 12/01/2023] Open
Abstract
Leukocytes possess the ability to migrate upstream-against the direction of flow-on surfaces of specific chemistry. Upstream migration was first characterized in vitro for T-cells on surfaces comprised of intracellular adhesion molecule-1 (ICAM-1). Upstream migration occurs when the integrin receptor αLβ2 (also known as lymphocyte function-associated antigen-1, or LFA-1) binds to ICAM-1. LFA-1/ICAM-1 interactions are ubiquitous and are widely found in leukocyte trafficking. Upstream migration would be employed after cells come to arrest on the apical surface of the endothelium and might confer an advantage for both trans-endothelial migration and tissue surveillance. It has now been shown that several other motile amoeboid cells which have the responsibility of trafficking from blood vessels into tissues, such as Marginal zone B cells, hematopoietic stem cells, and neutrophils (when macrophage-1 antigen, Mac-1, is blocked), can also migrate upstream on ICAM-1 surfaces. This review will summarize what is known about the basic mechanisms of upstream migration, which cells have displayed this phenomenon, and the possible role of upstream migration in physiology and tissue homeostasis.
Collapse
Affiliation(s)
- Alexander Buffone
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, United States
- Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, United States
| | - Daniel A. Hammer
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Sarah Hyun Ji Kim
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
| | | | - Ai Mochida
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Dong-Hun Lee
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Subham Guin
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, United States
| |
Collapse
|
19
|
González L, Mugler A. Collective effects in flow-driven cell migration. Phys Rev E 2023; 108:054406. [PMID: 38115469 DOI: 10.1103/physreve.108.054406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 10/17/2023] [Indexed: 12/21/2023]
Abstract
Autologous chemotaxis is the process in which cells secrete and detect molecules to determine the direction of fluid flow. Experiments and theory suggest that autologous chemotaxis fails at high cell densities because molecules from other cells interfere with a given cell's signal. We investigate autologous chemotaxis using a three-dimensional Monte Carlo-based motility simulation that couples spatial and temporal gradient sensing with cell-cell repulsion. Surprisingly, we find that when temporal gradient sensing dominates, high-density clusters chemotax faster than individual cells. To explain this observation, we propose a mechanism by which temporal gradient sensing allows cells to form a collective sensory unit. We demonstrate using computational fluid mechanics that that this mechanism indeed allows a cluster of cells to outperform single cells in terms of the detected anisotropy of the signal, a finding that we demonstrate with analytic scaling arguments. Our work suggests that collective autologous chemotaxis at high cell densities is possible and requires only known, ubiquitous cell capabilities.
Collapse
Affiliation(s)
- Louis González
- Department of Physics and Astronomy, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA
| | - Andrew Mugler
- Department of Physics and Astronomy, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA
| |
Collapse
|
20
|
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: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [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
|
21
|
Chen MB, Javanmardi Y, Shahreza S, Serwinski B, Aref A, Djordjevic B, Moeendarbary E. Mechanobiology in oncology: basic concepts and clinical prospects. Front Cell Dev Biol 2023; 11:1239749. [PMID: 38020912 PMCID: PMC10644154 DOI: 10.3389/fcell.2023.1239749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
The interplay between genetic transformations, biochemical communications, and physical interactions is crucial in cancer progression. Metastasis, a leading cause of cancer-related deaths, involves a series of steps, including invasion, intravasation, circulation survival, and extravasation. Mechanical alterations, such as changes in stiffness and morphology, play a significant role in all stages of cancer initiation and dissemination. Accordingly, a better understanding of cancer mechanobiology can help in the development of novel therapeutic strategies. Targeting the physical properties of tumours and their microenvironment presents opportunities for intervention. Advancements in imaging techniques and lab-on-a-chip systems enable personalized investigations of tumor biomechanics and drug screening. Investigation of the interplay between genetic, biochemical, and mechanical factors, which is of crucial importance in cancer progression, offers insights for personalized medicine and innovative treatment strategies.
Collapse
Affiliation(s)
- Michelle B. Chen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Yousef Javanmardi
- Department of Mechanical Engineering, University College London, London, United Kingdom
| | - Somayeh Shahreza
- Department of Mechanical Engineering, University College London, London, United Kingdom
| | - Bianca Serwinski
- Department of Mechanical Engineering, University College London, London, United Kingdom
- 199 Biotechnologies Ltd., London, United Kingdom
- Northeastern University London, London, United Kingdom
| | - Amir Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Boris Djordjevic
- Department of Mechanical Engineering, University College London, London, United Kingdom
- 199 Biotechnologies Ltd., London, United Kingdom
| | - Emad Moeendarbary
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Mechanical Engineering, University College London, London, United Kingdom
| |
Collapse
|
22
|
Bao L, Kong H, Ja Y, Wang C, Qin L, Sun H, Dai S. The relationship between cancer and biomechanics. Front Oncol 2023; 13:1273154. [PMID: 37901315 PMCID: PMC10602664 DOI: 10.3389/fonc.2023.1273154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/27/2023] [Indexed: 10/31/2023] Open
Abstract
The onset, development, diagnosis, and treatment of cancer involve intricate interactions among various factors, spanning the realms of mechanics, physics, chemistry, and biology. Within our bodies, cells are subject to a variety of forces such as gravity, magnetism, tension, compression, shear stress, and biological static force/hydrostatic pressure. These forces are perceived by mechanoreceptors as mechanical signals, which are then transmitted to cells through a process known as mechanical transduction. During tumor development, invasion and metastasis, there are significant biomechanical influences on various aspects such as tumor angiogenesis, interactions between tumor cells and the extracellular matrix (ECM), interactions between tumor cells and other cells, and interactions between tumor cells and the circulatory system and vasculature. The tumor microenvironment comprises a complex interplay of cells, ECM and vasculature, with the ECM, comprising collagen, fibronectins, integrins, laminins and matrix metalloproteinases, acting as a critical mediator of mechanical properties and a key component within the mechanical signaling pathway. The vasculature exerts appropriate shear forces on tumor cells, enabling their escape from immune surveillance, facilitating their dissemination in the bloodstream, dictating the trajectory of circulating tumor cells (CTCs) and playing a pivotal role in regulating adhesion to the vessel wall. Tumor biomechanics plays a critical role in tumor progression and metastasis, as alterations in biomechanical properties throughout the malignant transformation process trigger a cascade of changes in cellular behavior and the tumor microenvironment, ultimately culminating in the malignant biological behavior of the tumor.
Collapse
Affiliation(s)
- Liqi Bao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Renji College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hongru Kong
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yang Ja
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chengchao Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lei Qin
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hongwei Sun
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shengjie Dai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
23
|
Wang H, Lu J, Rathod M, Aw WY, Huang SA, Polacheck WJ. A facile fluid pressure system reveals differential cellular response to interstitial pressure gradients and flow. BIOMICROFLUIDICS 2023; 17:054103. [PMID: 37781136 PMCID: PMC10539030 DOI: 10.1063/5.0165119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/09/2023] [Indexed: 10/03/2023]
Abstract
Interstitial fluid pressure gradients and interstitial flow have been shown to drive morphogenic processes that shape tissues and influence progression of diseases including cancer. The advent of porous media microfluidic approaches has enabled investigation of the cellular response to interstitial flow, but questions remain as to the critical biophysical and biochemical signals imparted by interstitial fluid pressure gradients and resulting flow on resident cells and extracellular matrix (ECM). Here, we introduce a low-cost method to maintain physiological interstitial fluid pressures that is built from commonly accessible laboratory equipment, including a laser pointer, camera, Arduino board, and a commercially available linear actuator. We demonstrate that when the system is connected to a microfluidic device containing a 3D porous hydrogel, physiologic pressure is maintained with sub-Pascal resolution and when basic feedback control is directed using an Arduino, constant pressure and pressure gradient can be maintained even as cells remodel and degrade the ECM hydrogel over time. Using this model, we characterized breast cancer cell growth and ECM changes to ECM fibril structure and porosity in response to constant interstitial fluid pressure or constant interstitial flow. We observe increased collagen fibril bundling and the formation of porous structures in the vicinity of cancer cells in response to constant interstitial fluid pressure as compared to constant interstitial flow. Collectively, these results further define interstitial fluid pressure as a driver of key pathogenic responses in cells, and the systems and methods developed here will allow for future mechanistic work investigating mechanotransduction of interstitial fluid pressures and flows.
Collapse
Affiliation(s)
- Hao Wang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27514, USA
| | - Jingming Lu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27514, USA
| | - Mitesh Rathod
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27514, USA
| | - Wen Yih Aw
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27514, USA
| | - Stephanie A. Huang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27514, USA
| | | |
Collapse
|
24
|
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: 5] [Impact Index Per Article: 2.5] [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
|
25
|
Ren T, Maitusong M, Zhou X, Hong X, Cheng S, Lin Y, Xue J, Xu D, Chen J, Qian Y, Lu Y, Liu X, Zhu Y, Wang J. Programing Cell Assembly via Ink-Free, Label-Free Magneto-Archimedes Based Strategy. ACS NANO 2023; 17:12072-12086. [PMID: 37363813 DOI: 10.1021/acsnano.2c10704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Tissue engineering raised a high requirement to control cell distribution in defined materials and structures. In "ink"-based bioprintings, such as 3D printing and photolithography, cells were associated with inks for spatial orientation; the conditions suitable for one ink are hard to apply on other inks, which increases the obstacle in their universalization. The Magneto-Archimedes effect based (Mag-Arch) strategy can modulate cell locomotion directly without impelling inks. In a paramagnetic medium, cells were repelled from high magnetic strength zones due to their innate diamagnetism, which is independent of substrate properties. However, Mag-Arch has not been developed into a powerful bioprinting strategy as its precision, complexity, and throughput are limited by magnetic field distribution. By controlling the paramagnetic reagent concentration in the medium and the gaps between magnets, which decide the cell repelling scope of magnets, we created simultaneously more than a hundred micrometer scale identical assemblies into designed patterns (such as alphabets) with single/multiple cell types. Cell patterning models for cell migration and immune cell adhesion studies were conveniently created by Mag-Arch. As a proof of concept, we patterned a tumor/endothelial coculture model within a covered microfluidic channel to mimic epithelial-mesenchymal transition (EMT) under shear stress in a cancer pathological environment, which gave a potential solution to pattern multiple cell types in a confined space without any premodification. Overall, our Mag-Arch patterning presents an alternative strategy for the biofabrication and biohybrid assembly of cells with biomaterials featured in controlled distribution and organization, which can be broadly employed in tissue engineering, regenerative medicine, and cell biology research.
Collapse
Affiliation(s)
- Tanchen Ren
- Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310029, P.R. China
| | - Miribani Maitusong
- Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310029, P.R. China
| | - Xuhao Zhou
- Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310029, P.R. China
| | - Xiaoqian Hong
- Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310029, P.R. China
| | - Si Cheng
- Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310029, P.R. China
| | - Yin Lin
- Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310029, P.R. China
| | - Junhui Xue
- Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310029, P.R. China
| | - Dilin Xu
- Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310029, P.R. China
| | - Jinyong Chen
- Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310029, P.R. China
| | - Yi Qian
- Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310029, P.R. China
| | - Yuwen Lu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, P.R. China
| | - Xianbao Liu
- Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310029, P.R. China
| | - Yang Zhu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, P.R. China
| | - Jian'an Wang
- Department of Cardiology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310029, P.R. China
| |
Collapse
|
26
|
Saraswathibhatla A, Indana D, Chaudhuri O. Cell-extracellular matrix mechanotransduction in 3D. Nat Rev Mol Cell Biol 2023; 24:495-516. [PMID: 36849594 PMCID: PMC10656994 DOI: 10.1038/s41580-023-00583-1] [Citation(s) in RCA: 175] [Impact Index Per Article: 87.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2023] [Indexed: 03/01/2023]
Abstract
Mechanical properties of extracellular matrices (ECMs) regulate essential cell behaviours, including differentiation, migration and proliferation, through mechanotransduction. Studies of cell-ECM mechanotransduction have largely focused on cells cultured in 2D, on top of elastic substrates with a range of stiffnesses. However, cells often interact with ECMs in vivo in a 3D context, and cell-ECM interactions and mechanisms of mechanotransduction in 3D can differ from those in 2D. The ECM exhibits various structural features as well as complex mechanical properties. In 3D, mechanical confinement by the surrounding ECM restricts changes in cell volume and cell shape but allows cells to generate force on the matrix by extending protrusions and regulating cell volume as well as through actomyosin-based contractility. Furthermore, cell-matrix interactions are dynamic owing to matrix remodelling. Accordingly, ECM stiffness, viscoelasticity and degradability often play a critical role in regulating cell behaviours in 3D. Mechanisms of 3D mechanotransduction include traditional integrin-mediated pathways that sense mechanical properties and more recently described mechanosensitive ion channel-mediated pathways that sense 3D confinement, with both converging on the nucleus for downstream control of transcription and phenotype. Mechanotransduction is involved in tissues from development to cancer and is being increasingly harnessed towards mechanotherapy. Here we discuss recent progress in our understanding of cell-ECM mechanotransduction in 3D.
Collapse
Affiliation(s)
| | - Dhiraj Indana
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA.
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA.
| |
Collapse
|
27
|
Moon HR, Saha S, Mugler A, Han B. Cells function as a ternary logic gate to decide migration direction under integrated chemical and fluidic cues. LAB ON A CHIP 2023; 23:631-644. [PMID: 36524874 PMCID: PMC9926949 DOI: 10.1039/d2lc00807f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/22/2022] [Indexed: 06/17/2023]
Abstract
Cells sense various environmental cues and subsequently process intracellular signals to decide their migration direction in many physiological and pathological processes. Although several signaling molecules and networks have been identified in these directed migrations, it still remains ambiguous to predict the migration direction under multiple and integrated cues, specifically chemical and fluidic cues. Here, we investigated the cellular signal processing machinery by reverse-engineering directed cell migration under integrated chemical and fluidic cues. We imposed controlled chemical and fluidic cues to cells using a microfluidic platform and analyzed the extracellular coupling of the cues with respect to the cellular detection limit. Then, the cell's migratory behavior was reverse-engineered to build a cellular signal processing system as a logic gate, which is based on a "selection" gate. This framework is further discussed with a minimal intracellular signaling network of a shared pathway model. The proposed framework of the ternary logic gate suggests a systematic view to understand how cells decode multiple cues and make decisions about the migration direction.
Collapse
Affiliation(s)
- Hye-Ran Moon
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA.
| | - Soutick Saha
- Department of Physics and Astronomy, Purdue University, West Lafayette, IN, USA
| | - Andrew Mugler
- Department of Physics and Astronomy, Purdue University, West Lafayette, IN, USA
- Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, USA
- Department of Physics and Astronomy, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Bumsoo Han
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA.
- Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
28
|
Barrasa-Ramos S, Dessalles CA, Hautefeuille M, Barakat AI. Mechanical regulation of the early stages of angiogenesis. J R Soc Interface 2022; 19:20220360. [PMID: 36475392 PMCID: PMC9727679 DOI: 10.1098/rsif.2022.0360] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Favouring or thwarting the development of a vascular network is essential in fields as diverse as oncology, cardiovascular disease or tissue engineering. As a result, understanding and controlling angiogenesis has become a major scientific challenge. Mechanical factors play a fundamental role in angiogenesis and can potentially be exploited for optimizing the architecture of the resulting vascular network. Largely focusing on in vitro systems but also supported by some in vivo evidence, the aim of this Highlight Review is dual. First, we describe the current knowledge with particular focus on the effects of fluid and solid mechanical stimuli on the early stages of the angiogenic process, most notably the destabilization of existing vessels and the initiation and elongation of new vessels. Second, we explore inherent difficulties in the field and propose future perspectives on the use of in vitro and physics-based modelling to overcome these difficulties.
Collapse
Affiliation(s)
- Sara Barrasa-Ramos
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Claire A. Dessalles
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Mathieu Hautefeuille
- Laboratoire de Biologie du Développement (UMR7622), Institut de Biologie Paris Seine, Sorbonne Université, Paris, France,Facultad de Ciencias, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Abdul I. Barakat
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| |
Collapse
|
29
|
Keratocytes migrate against flow with a roly-poly-like mechanism. Proc Natl Acad Sci U S A 2022; 119:e2210379119. [PMID: 36409912 PMCID: PMC9889884 DOI: 10.1073/pnas.2210379119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
While cell migration can be directed by various mechanical cues such as force, deformation, stiffness, or flow, the associated mechanisms and functions may remain elusive. Single cell migration against flow, repeatedly reported with leukocytes, is arguably considered as active and mediated by integrin mechanotransduction, or passive and determined by a mechanical bias. Here, we reveal a phenotype of flow mechanotaxis with fish epithelial keratocytes that orient upstream or downstream at shear stresses around tens of dyn cm-2. We show that each cell has an intrinsic orientation that results from the mechanical interaction of flow with its morphology. The bulbous trailing edge of a keratocyte generates a hydrodynamical torque under flow that stabilizes an upstream orientation, just as the heavy lower edge of a roly-poly toy generates a gravitational torque that stabilizes an upright position. In turn, the wide and flat leading edge of keratocytes destabilizes upstream orientation, allowing the existence of two distinct phenotypes. To formalize these observations, we propose a simple mechanical model that considers keratocyte morphology as a hemisphere preceded by a wide thin sheet. Our findings show that this model can recapitulate the phase diagram of single cell orientation under flow without adjustable parameters. From a larger perspective, this passive mechanism of keratocytes flow mechanotaxis implies a potential absence of physiological function and evolution-driven process.
Collapse
|
30
|
Imashiro C, Mei J, Friend J, Takemura K. Quantifying cell adhesion through forces generated by acoustic streaming. ULTRASONICS SONOCHEMISTRY 2022; 90:106204. [PMID: 36257212 PMCID: PMC9583098 DOI: 10.1016/j.ultsonch.2022.106204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/01/2022] [Accepted: 10/12/2022] [Indexed: 06/16/2023]
Abstract
The strength of cell adhesion is important in understanding the cell's health and in culturing them. Quantitative measurement of cell adhesion strength is a significant challenge in bioengineering research. For this, the present study describes a system that can measure cell adhesion strength using acoustic streaming induced by Lamb waves. Cells are cultured on an ultrasound transducer using a range of preculture and incubation times with phosphate-buffered saline (PBS) just before the measurement. Acoustic streaming is then induced using several Lamb wave intensities, exposing the cells to shear flows and eventually detaching them. By relying upon a median detachment rate of 50 %, the corresponding detachment force, or force of cell adhesion, was determined to be on the order of several nN, consistent with previous reports. The stronger the induced shear flow, the more cells were detached. Further, we employed a preculture time of 8 to 24 h and a PBS incubation time of 0 to 60 min, producing cell adhesion forces that varied from 1.2 to 13 nN. Hence, the developed system can quantify cell adhesion strength over a wide range, possibly offering a fundamental tool for cell-based bioengineering.
Collapse
Affiliation(s)
- Chikahiro Imashiro
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan; Department of Mechanical Engineering, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa 223-8522, Japan.
| | - Jiyang Mei
- Medically Advanced Devices Laboratory, Center for Medical Devices, Department of Mechanical and Aerospace Engineering, Jacobs School of Engineering and Department of Surgery, School of Medicine, University of California, San Diego, CA 92093, USA
| | - James Friend
- Medically Advanced Devices Laboratory, Center for Medical Devices, Department of Mechanical and Aerospace Engineering, Jacobs School of Engineering and Department of Surgery, School of Medicine, University of California, San Diego, CA 92093, USA
| | - Kenjiro Takemura
- Department of Mechanical Engineering, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa 223-8522, Japan
| |
Collapse
|
31
|
Wang J, Han Y, Li Y, Zhang F, Cai M, Zhang X, Chen J, Ji C, Ma J, Xu F. Targeting Tumor Physical Microenvironment for Improved Radiotherapy. SMALL METHODS 2022; 6:e2200570. [PMID: 36116123 DOI: 10.1002/smtd.202200570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/23/2022] [Indexed: 06/15/2023]
Abstract
Radiotherapy has led to important clinical advances; existing cancer radiotherapy resistance is one remaining major challenge. Recently, biophysical cues in the tumor microenvironment (TME) have been regarded as the new hallmarks of cancer, playing pivotal roles in various cancer behaviors and treatment responses, including radiotherapy resistance. With recent advances in micro/nanotechnologies and functional biomaterials, radiotherapy exerts great influence on biophysical cues in TME, which, in turn, significantly affect the response to radiotherapy. Besides, various strategies have emerged that target biophysical cues in TME, to potentially enhance radiotherapy efficacy. Therefore, this paper reviews the four biophysical cues (i.e., extracellular matrix (ECM) microarchitecture, ECM stiffness, interstitial fluid pressure, and solid stress) that may play important roles in radiotherapy resistance, their possible mechanisms for inducing it, and their change after radiotherapy. The emerging therapeutic strategies targeting the biophysical microenvironment, to explore the mechanism of radiotherapy resistance and develop effective strategies to revert it for improved treatment efficacy are further summarized.
Collapse
Affiliation(s)
- Jin Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Yulong Han
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, P. R. China
| | - Yuan Li
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Fengping Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Mengjiao Cai
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Xinyue Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Jie Chen
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Chao Ji
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Jinlu Ma
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| |
Collapse
|
32
|
Guimarães CF, Marques AP, Reis RL. Pushing the Natural Frontier: Progress on the Integration of Biomaterial Cues toward Combinatorial Biofabrication and Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2105645. [PMID: 35419887 DOI: 10.1002/adma.202105645] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 03/14/2022] [Indexed: 06/14/2023]
Abstract
The engineering of fully functional, biological-like tissues requires biomaterials to direct cellular events to a near-native, 3D niche extent. Natural biomaterials are generally seen as a safe option for cell support, but their biocompatibility and biodegradability can be just as limited as their bioactive/biomimetic performance. Furthermore, integrating different biomaterial cues and their final impact on cellular behavior is a complex equation where the outcome might be very different from the sum of individual parts. This review critically analyses recent progress on biomaterial-induced cellular responses, from simple adhesion to more complex stem cell differentiation, looking at the ever-growing possibilities of natural materials modification. Starting with a discussion on native material formulation and the inclusion of cell-instructive cues, the roles of shape and mechanical stimuli, the susceptibility to cellular remodeling, and the often-overlooked impact of cellular density and cell-cell interactions within constructs, are delved into. Along the way, synergistic and antagonistic combinations reported in vitro and in vivo are singled out, identifying needs and current lessons on the development of natural biomaterial libraries to solve the cell-material puzzle efficiently. This review brings together knowledge from different fields envisioning next-generation, combinatorial biomaterial development toward complex tissue engineering.
Collapse
Affiliation(s)
- Carlos F Guimarães
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Alexandra P Marques
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
33
|
Jones CFE, Di Cio S, Connelly JT, Gautrot JE. Design of an Integrated Microvascularized Human Skin-on-a-Chip Tissue Equivalent Model. Front Bioeng Biotechnol 2022; 10:915702. [PMID: 35928950 PMCID: PMC9343775 DOI: 10.3389/fbioe.2022.915702] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Tissue-engineered skin constructs have been under development since the 1980s as a replacement for human skin tissues and animal models for therapeutics and cosmetic testing. These have evolved from simple single-cell assays to increasingly complex models with integrated dermal equivalents and multiple cell types including a dermis, epidermis, and vasculature. The development of micro-engineered platforms and biomaterials has enabled scientists to better recreate and capture the tissue microenvironment in vitro, including the vascularization of tissue models and their integration into microfluidic chips. However, to date, microvascularized human skin equivalents in a microfluidic context have not been reported. Here, we present the design of a novel skin-on-a-chip model integrating human-derived primary and immortalized cells in a full-thickness skin equivalent. The model is housed in a microfluidic device, in which a microvasculature was previously established. We characterize the impact of our chip design on the quality of the microvascular networks formed and evidence that this enables the formation of more homogenous networks. We developed a methodology to harvest tissues from embedded chips, after 14 days of culture, and characterize the impact of culture conditions and vascularization (including with pericyte co-cultures) on the stratification of the epidermis in the resulting skin equivalents. Our results indicate that vascularization enhances stratification and differentiation (thickness, architecture, and expression of terminal differentiation markers such as involucrin and transglutaminase 1), allowing the formation of more mature skin equivalents in microfluidic chips. The skin-on-a-chip tissue equivalents developed, because of their realistic microvasculature, may find applications for testing efficacy and safety of therapeutics delivered systemically, in a human context.
Collapse
Affiliation(s)
- Christian F. E. Jones
- Institute of Bioengineering, Queen Mary University of London, London, United Kingdom
- School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| | - Stefania Di Cio
- Institute of Bioengineering, Queen Mary University of London, London, United Kingdom
- School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| | - John T. Connelly
- The Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Julien E. Gautrot
- Institute of Bioengineering, Queen Mary University of London, London, United Kingdom
- School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
34
|
Grigoryan EN. Self-Organization of the Retina during Eye Development, Retinal Regeneration In Vivo, and in Retinal 3D Organoids In Vitro. Biomedicines 2022; 10:1458. [PMID: 35740479 PMCID: PMC9221005 DOI: 10.3390/biomedicines10061458] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/16/2022] [Accepted: 06/18/2022] [Indexed: 11/23/2022] Open
Abstract
Self-organization is a process that ensures histogenesis of the eye retina. This highly intricate phenomenon is not sufficiently studied due to its biological complexity and genetic heterogeneity. The review aims to summarize the existing central theories and ideas for a better understanding of retinal self-organization, as well as to address various practical problems of retinal biomedicine. The phenomenon of self-organization is discussed in the spatiotemporal context and illustrated by key findings during vertebrate retina development in vivo and retinal regeneration in amphibians in situ. Described also are histotypic 3D structures obtained from the disaggregated retinal progenitor cells of birds and retinal 3D organoids derived from the mouse and human pluripotent stem cells. The review highlights integral parts of retinal development in these conditions. On the cellular level, these include competence, differentiation, proliferation, apoptosis, cooperative movements, and migration. On the physical level, the focus is on the mechanical properties of cell- and cell layer-derived forces and on the molecular level on factors responsible for gene regulation, such as transcription factors, signaling molecules, and epigenetic changes. Finally, the self-organization phenomenon is discussed as a basis for the production of retinal organoids, a promising model for a wide range of basic scientific and medical applications.
Collapse
Affiliation(s)
- Eleonora N Grigoryan
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| |
Collapse
|
35
|
Mehta P, Rahman Z, Ten Dijke P, Boukany PE. Microfluidics meets 3D cancer cell migration. Trends Cancer 2022; 8:683-697. [PMID: 35568647 DOI: 10.1016/j.trecan.2022.03.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 03/23/2022] [Accepted: 03/30/2022] [Indexed: 01/12/2023]
Abstract
An early step of metastasis requires a complex and coordinated migration of invasive tumor cells into the surrounding tumor microenvironment (TME), which contains extracellular matrix (ECM). It is being appreciated that 3D matrix-based microfluidic models have an advantage over conventional in vitro and animal models to study tumor progression events. Recent microfluidic models have enabled recapitulation of key mechanobiological features present within the TME to investigate collective cancer cell migration and invasion. Microfluidics also allows for functional interrogation and therapeutic manipulation of specific steps to study the dynamic aspects of tumor progression. In this review, we focus on recent developments in cancer cell migration and how microfluidic strategies have evolved to address the physiological complexities of the TME to visualize migration modes adapted by various tumor cells.
Collapse
Affiliation(s)
- Pranav Mehta
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands; Department of Chemical Engineering, Delft University of Technology, Van der Maasweg 9, 2629 HZ, Delft, The Netherlands
| | - Zaid Rahman
- Department of Chemical Engineering, Delft University of Technology, Van der Maasweg 9, 2629 HZ, Delft, The Netherlands
| | - Peter Ten Dijke
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands.
| | - Pouyan E Boukany
- Department of Chemical Engineering, Delft University of Technology, Van der Maasweg 9, 2629 HZ, Delft, The Netherlands.
| |
Collapse
|
36
|
Xu H, Cheng C, Le W. Recent research advances of the biomimetic tumor microenvironment and regulatory factors on microfluidic devices: A systematic review. Electrophoresis 2022; 43:839-847. [PMID: 35179796 DOI: 10.1002/elps.202100360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 02/08/2022] [Accepted: 02/11/2022] [Indexed: 11/07/2022]
Abstract
Tumor microenvironment is a multicomponent system consisting of tumor cells, noncancer cells, extracellular matrix, and signaling molecules, which hosts tumor cells with integrated biophysical and biochemical elements. Because of its critical involvement in tumor genesis, invasion, metastasis, and resistance, the tumor microenvironment is emerging as a hot topic of tumor biology and a prospective therapeutic target. Unfortunately, the complex of microenvironment modeling in vitro is technically challenging and does not effectively generalize the local tumor tissue milieu. Recently, significant advances in microfluidic technologies have provided us with an approach to imitate physiological systems that can be utilized to mimic the characterization of tumor responses with pathophysiological relevance in vitro. In this review, we highlight the recent progress and innovations in microfluidic technology that facilitates the tumor microenvironment study. We also discuss the progress and future perspective of microfluidic bionic approaches with high efficiency for the study of tumor microenvironment and the challenges encountered in cancer research, drug discovery, and personalized therapy.
Collapse
Affiliation(s)
- Hui Xu
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, P. R. China
| | - Cheng Cheng
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, P. R. China
| | - Weidong Le
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, P. R. China.,Institute of Neurology, Sichuan Academy of Medical Sciences-Sichuan Provincial People's Hospital, Chengdu, P. R. China
| |
Collapse
|
37
|
Beeghly GF, Amofa KY, Fischbach C, Kumar S. Regulation of Tumor Invasion by the Physical Microenvironment: Lessons from Breast and Brain Cancer. Annu Rev Biomed Eng 2022; 24:29-59. [PMID: 35119915 DOI: 10.1146/annurev-bioeng-110220-115419] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The success of anticancer therapies is often limited by heterogeneity within and between tumors. While much attention has been devoted to understanding the intrinsic molecular diversity of tumor cells, the surrounding tissue microenvironment is also highly complex and coevolves with tumor cells to drive clinical outcomes. Here, we propose that diverse types of solid tumors share common physical motifs that change in time and space, serving as universal regulators of malignancy. We use breast cancer and glioblastoma as instructive examples and highlight how invasion in both diseases is driven by the appropriation of structural guidance cues, contact-dependent heterotypic interactions with stromal cells, and elevated interstitial fluid pressure and flow. We discuss how engineering strategies show increasing value for measuring and modeling these physical properties for mechanistic studies. Moreover, engineered systems offer great promise for developing and testing novel therapies that improve patient prognosis by normalizing the physical tumor microenvironment. Expected final online publication date for the Annual Review of Biomedical Engineering, Volume 24 is June 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Garrett F Beeghly
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA;
| | - Kwasi Y Amofa
- University of California, Berkeley-University of California, San Francisco Graduate Program in Bioengineering, Berkeley, California, USA; .,Department of Bioengineering, University of California, Berkeley, Berkeley, California, USA
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA; .,Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, New York, USA
| | - Sanjay Kumar
- University of California, Berkeley-University of California, San Francisco Graduate Program in Bioengineering, Berkeley, California, USA; .,Department of Bioengineering, University of California, Berkeley, Berkeley, California, USA.,Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California, USA.,Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
38
|
Nath S, Pigula M, Hasan T, Rizvi I. A Perfusion Model to Evaluate Response to Photodynamic Therapy in 3D Tumors. Methods Mol Biol 2022; 2451:49-58. [PMID: 35505009 DOI: 10.1007/978-1-0716-2099-1_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Numerous cancer models have been developed to investigate the effects of mechanical stress on the biology of cells. Here we describe a protocol to fabricate a perfusion model to culture 3-dimensional (3D) ovarian cancer nodules under constant flow. The modular design of this model allows for a wide range of treatment regimens and combinations, including PDT and chemotherapy. Finally, methods for a number of readouts are detailed, allowing researchers to investigate a variety of biological and cytotoxic parameters related to mechanical stress and therapeutic modalities.
Collapse
Affiliation(s)
- Shubhankar Nath
- Wellman Center for Photomedicine, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Michael Pigula
- Wellman Center for Photomedicine, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
- Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Imran Rizvi
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
39
|
López-Alonso I, López-Martínez C, Martín-Vicente P, Amado-Rodríguez L, González-López A, Mayordomo-Colunga J, Del Busto C, Bernal M, Crespo I, Astudillo A, Arias-Guillén M, Fueyo A, Almendros I, Otero J, Sanz-Fraile H, Farré R, Albaiceta GM. Mechanical ventilation promotes lung tumor spread by modulation of cholesterol cell content. Eur Respir J 2021; 60:13993003.01470-2021. [PMID: 34887328 DOI: 10.1183/13993003.01470-2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 11/22/2021] [Indexed: 11/05/2022]
Abstract
Mechanical stretch of cancer cells can alter their invasiveness. During mechanical ventilation, lungs may be exposed to an increased amount of stretch, but the consequences on lung tumors have not been explored. To characterize the influence of mechanical ventilation on the behavior of lung tumors, invasiveness assays and transcriptomic analyses were performed in cancer cell lines cultured in static conditions or under cyclic stretch. Mice harbouring lung melanoma implants were submitted to mechanical ventilation and metastatic spread was assessed. Additional in vivo experiments were performed to determine the mechano-dependent specificity of the response. Incidence of metastases was studied in a cohort of lung cancer patients that received mechanical ventilation compared with a matched group of non-ventilated patients. Stretch increases invasiveness in melanoma B16F10luc2 and lung adenocarcinoma A549 cells. We identified a mechanosensitive upregulation of pathways involved in cholesterol processing in vitro, leading to an increase in PCSK9 and LDLR expression, a decrease in intracellular cholesterol and preservation of cell stiffness. A course of mechanical ventilation in mice harboring melanoma implants increased brain and kidney metastases two weeks later. Blockade of PCSK9 using a monoclonal antibody increased cell cholesterol and stiffness and decreased cell invasiveness in vitro and metastasis in vivo In patients, mechanical ventilation increased PCSK9 abundance in lung tumors and the incidence of metastasis, thus decreasing survival. Our results suggest that mechanical stretch promote invasiveness of cancer cells, which may have clinically relevant consequences. Pharmacological manipulation of cholesterol endocytosis could be a novel therapeutic target in this setting.
Collapse
Affiliation(s)
- Inés López-Alonso
- Centro de Investigación Biomédica en Red-Enfermedades Respiratorias, Madrid, Spain .,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain.,These authors contributed equally
| | - Cecilia López-Martínez
- Centro de Investigación Biomédica en Red-Enfermedades Respiratorias, Madrid, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,These authors contributed equally
| | - Paula Martín-Vicente
- Centro de Investigación Biomédica en Red-Enfermedades Respiratorias, Madrid, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Laura Amado-Rodríguez
- Centro de Investigación Biomédica en Red-Enfermedades Respiratorias, Madrid, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain.,Unidad de Cuidados Intensivos Cardiológicos. Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Adrián González-López
- Centro de Investigación Biomédica en Red-Enfermedades Respiratorias, Madrid, Spain.,Department of Anesthesiology and Operative Intensive Care Medicine CCM/CVK, Charité - Universitätsmedizin Berlin, Germany
| | - Juan Mayordomo-Colunga
- Centro de Investigación Biomédica en Red-Enfermedades Respiratorias, Madrid, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,Unidad de Cuidados Intensivos Pediátricos. Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Cecilia Del Busto
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,Unidad de Cuidados Intensivos Polivalente. Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Marina Bernal
- Servicio de Medicina Interna. Fundación Jiménez Díaz, Madrid, Spain
| | - Irene Crespo
- Departamento de Biología Funcional, Universidad de Oviedo, Oviedo, Spain
| | - Aurora Astudillo
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Miguel Arias-Guillén
- Centro de Investigación Biomédica en Red-Enfermedades Respiratorias, Madrid, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,Servicio de Neumología. Hospital Unviersitario Central de Asturias. Oviedo, Spain
| | - Antonio Fueyo
- Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain.,Departamento de Biología Funcional, Universidad de Oviedo, Oviedo, Spain.,Centro de Investigación Biomédica en Red-Oncología, Madrid, Spain
| | - Isaac Almendros
- Centro de Investigación Biomédica en Red-Enfermedades Respiratorias, Madrid, Spain.,Unitat Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.,Institut Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain
| | - Jorge Otero
- Centro de Investigación Biomédica en Red-Enfermedades Respiratorias, Madrid, Spain.,Unitat Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Héctor Sanz-Fraile
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Ramón Farré
- Centro de Investigación Biomédica en Red-Enfermedades Respiratorias, Madrid, Spain.,Unitat Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.,Institut Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain
| | - Guillermo M Albaiceta
- Centro de Investigación Biomédica en Red-Enfermedades Respiratorias, Madrid, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain.,Unidad de Cuidados Intensivos Cardiológicos. Hospital Universitario Central de Asturias, Oviedo, Spain.,Departamento de Biología Funcional, Universidad de Oviedo, Oviedo, Spain
| |
Collapse
|
40
|
Chonan Y, Yamashita T, Sampetrean O, Saya H, Sudo R. Spatial heterogeneity of invading glioblastoma cells regulated by paracrine factors. Tissue Eng Part A 2021; 28:573-585. [PMID: 34841881 DOI: 10.1089/ten.tea.2021.0168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma (GBM) is the most common and lethal type of malignant primary brain tumor in adults. GBM displays heterogeneous tumor cell population comprising glioma-initiating cells (GICs) with stem cell-like characteristics and differentiated glioma cells. During GBM cell invasion into normal brain tissues, which is the hallmark characteristic of GBM, GICs at the invasion front retain stemness, while cells at the tumor core display cellular differentiation. However, the mechanism of cellular differentiation underlying the formation of spatial cellular heterogeneity in GBM remains unknown. In the present study, we first observed spatially heterogeneous GBM cell populations emerged from an isogenic clonal population of GICs during invasion into a 3D collagen hydrogel in a microfluidic device. Specifically, GICs at the invasion front maintained stemness, while trailing cells displayed astrocytic differentiation. The spatial cellular heterogeneity resulted from the difference in cell density between GICs at the invasion front and trailing cells. Trailing GICs at high cell density exhibited astrocytic differentiation via local accumulation of paracrine factors they secreted, while cells at the invasion front of low cell density retained stemness due to the lack of paracrine factors. In addition, we demonstrated that interstitial flow suppressed astrocytic differentiation of trailing GICs by the clearance of paracrine factors. Our findings suggest that intercellular crosstalk between tumor cells is an essential factor in developing the spatial cellular heterogeneity of GBM cells with various differentiation statuses. It also provides insights into the development of novel therapeutic strategies targeting GBM cells with stem cell characteristics at the invasion front.
Collapse
Affiliation(s)
- Yuta Chonan
- Keio University, School of Integrated Design Engineering, Yokohama, Kanagawa, Japan;
| | - Tadahiro Yamashita
- Keio University, Department of System Design Engineering, Yokohama, Kanagawa, Japan.,Keio University, School of Integrated Design Engineering, Yokohama, Kanagawa, Japan;
| | - Oltea Sampetrean
- Keio University School of Medicine, Division of Gene Regulation, Institute for Advanced Medical Research, Tokyo, Japan;
| | - Hideyuki Saya
- Keio University School of Medicine, Division of Gene Regulation, Institute for Advanced Medical Research, Tokyo, Japan;
| | - Ryo Sudo
- Keio University, Department of System Design Engineering, Yokohama, Kanagawa, Japan.,Keio University, School of Integrated Design Engineering, Yokohama, Kanagawa, Japan;
| |
Collapse
|
41
|
Ravichandran A, Clegg J, Adams MN, Hampson M, Fielding A, Bray LJ. 3D Breast Tumor Models for Radiobiology Applications. Cancers (Basel) 2021; 13:5714. [PMID: 34830869 PMCID: PMC8616164 DOI: 10.3390/cancers13225714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/28/2021] [Accepted: 11/07/2021] [Indexed: 12/17/2022] Open
Abstract
Breast cancer is a leading cause of cancer-associated death in women. The clinical management of breast cancers is normally carried out using a combination of chemotherapy, surgery and radiation therapy. The majority of research investigating breast cancer therapy until now has mainly utilized two-dimensional (2D) in vitro cultures or murine models of disease. However, there has been significant uptake of three-dimensional (3D) in vitro models by cancer researchers over the past decade, highlighting a complimentary model for studies of radiotherapy, especially in conjunction with chemotherapy. In this review, we underline the effects of radiation therapy on normal and malignant breast cells and tissues, and explore the emerging opportunities that pre-clinical 3D models offer in improving our understanding of this treatment modality.
Collapse
Affiliation(s)
- Akhilandeshwari Ravichandran
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; (A.R.); (J.C.); (M.H.)
- ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia;
| | - Julien Clegg
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; (A.R.); (J.C.); (M.H.)
- ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia;
| | - Mark N. Adams
- ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia;
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Madison Hampson
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; (A.R.); (J.C.); (M.H.)
| | - Andrew Fielding
- School of Chemistry and Physics, Queensland University of Technology, Brisbane, QLD 4000, Australia;
| | - Laura J. Bray
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; (A.R.); (J.C.); (M.H.)
- ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia;
| |
Collapse
|
42
|
Haase K, Piatti F, Marcano M, Shin Y, Visone R, Redaelli A, Rasponi M, Kamm RD. Physiologic flow-conditioning limits vascular dysfunction in engineered human capillaries. Biomaterials 2021; 280:121248. [PMID: 34794827 DOI: 10.1016/j.biomaterials.2021.121248] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 02/02/2023]
Abstract
Hemodynamics play a central role in the health and disease of the coronary and peripheral vascular systems. Vessel-lining endothelial cells are known mechanosensors, responding to disturbances in flow - with mechanosensitivity hypothesized to change in response to metabolic demands. The health of our smallest microvessels have been lauded as a prognostic marker for cardiovascular health. Yet, despite numerous animal models, studying these small vessels has proved difficult. Microfluidic technologies have allowed a number of 3D vascular models to be developed and used to investigate human vessels. Here, two such systems are employed for examining 1) interstitial flow effects on neo-vessel formation, and 2) the effects of flow-conditioning on vascular remodeling following sustained static culture. Interstitial flow is shown to enhance early vessel formation via significant remodeling of vessels and interconnected tight junctions of the endothelium. In formed vessels, continuous flow maintains a stable vascular diameter and causes significant remodeling, contrasting the continued anti-angiogenic decline of statically cultured vessels. This study is the first to couple complex 3D computational flow distributions and microvessel remodeling from microvessels grown on-chip (exposed to flow or no-flow conditions). Flow-conditioned vessels (WSS < 1Pa for 30 μm vessels) increase endothelial barrier function, result in significant changes in gene expression and reduce reactive oxygen species and anti-angiogenic cytokines. Taken together, these results demonstrate microvessel mechanosensitivity to flow-conditioning, which limits deleterious vessel regression in vitro, and could have implications for future modeling of reperfusion/no-flow conditions.
Collapse
Affiliation(s)
- Kristina Haase
- Dept. of Mechanical Engineering, MIT, Cambridge, MA, USA
| | - Filippo Piatti
- Dept. of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| | | | - Yoojin Shin
- Dept. of Mechanical Engineering, MIT, Cambridge, MA, USA
| | - Roberta Visone
- Dept. of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Alberto Redaelli
- Dept. of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Marco Rasponi
- Dept. of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Roger D Kamm
- Dept. of Mechanical Engineering, MIT, Cambridge, MA, USA; Dept. of Biological Engineering, MIT, Cambridge, MA, USA.
| |
Collapse
|
43
|
Shirke PU, Goswami H, Kumar V, Shah D, Beri S, Das S, Bellare J, Mayor S, Venkatesh KV, Seth JR, Majumder A. "Viscotaxis"- directed migration of mesenchymal stem cells in response to loss modulus gradient. Acta Biomater 2021; 135:356-367. [PMID: 34469788 PMCID: PMC7616456 DOI: 10.1016/j.actbio.2021.08.039] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 07/30/2021] [Accepted: 08/24/2021] [Indexed: 12/12/2022]
Abstract
Directed cell migration plays a crucial role in physiological and pathological conditions. One important mechanical cue, known to influence cell migration, is the gradient of substrate elastic modulus (E). However, the cellular microenvironment is viscoelastic and hence the elastic property alone is not sufficient to define its material characteristics. To bridge this gap, in this study, we investigated the influence of the gradient of viscous property of the substrate, as defined by loss modulus (G″) on cell migration. We cultured human mesenchymal stem cells (hMSCs) on a collagen-coated polyacrylamide gel with constant storage modulus (G') but with a gradient in the loss modulus (G″). We found hMSCs to migrate from high to low loss modulus. We have termed this form of directional cellular migration as "Viscotaxis". We hypothesize that the high loss modulus regime deforms more due to creep in the long timescale when subjected to cellular traction. Such differential deformation drives the observed Viscotaxis. To verify our hypothesis, we disrupted the actomyosin contractility with myosin inhibitor blebbistatin and ROCK inhibitor Y27632, and found the directional migration to disappear. Further, such time-dependent creep of the high loss material should lead to lower traction, shorter lifetime of the focal adhesions, and dynamic cell morphology, which was indeed found to be the case. Together, findings in this paper highlight the importance of considering the viscous modulus while preparing stiffness-based substrates for the field of tissue engineering. STATEMENT OF SIGNIFICANCE: While the effect of substrate elastic modulus has been investigated extensively in the context of cell biology, the role of substrate viscoelasticity is poorly understood. This omission is surprising as our body is not elastic, but viscoelastic. Hence, the role of viscoelasticity needs to be investigated at depth in various cellular contexts. One such important context is cell migration. Cell migration is important in morphogenesis, immune response, wound healing, and cancer, to name a few. While it is known that cells migrate when presented with a substrate with a rigidity gradient, cellular behavior in response to viscoelastic gradient has never been investigated. The findings of this paper not only reveal a completely novel cellular taxis or directed migration, it also improves our understanding of cell mechanics significantly.
Collapse
Affiliation(s)
- Pallavi Uday Shirke
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Hiya Goswami
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Vardhman Kumar
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Darshan Shah
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Sarayu Beri
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bellary Road, Bangalore, India
| | - Siddhartha Das
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Jayesh Bellare
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Satyajit Mayor
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bellary Road, Bangalore, India
| | - K V Venkatesh
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Jyoti R Seth
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India.
| | - Abhijit Majumder
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India.
| |
Collapse
|
44
|
Hao J, Zhao W, Oh JM, Shen K. A Pillar-Free Diffusion Device for Studying Chemotaxis on Supported Lipid Bilayers. MICROMACHINES 2021; 12:mi12101254. [PMID: 34683305 PMCID: PMC8538285 DOI: 10.3390/mi12101254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/15/2021] [Accepted: 10/15/2021] [Indexed: 11/21/2022]
Abstract
Chemotactic cell migration plays a crucial role in physiological and pathophysiological processes. In tissues, cells can migrate not only through extracellular matrix (ECM), but also along stromal cell surfaces via membrane-bound receptor–ligand interactions to fulfill critical functions. However, there remains a lack of models recapitulating chemotactic migration mediated through membrane-bound interactions. Here, using micro-milling, we engineered a multichannel diffusion device that incorporates a chemoattractant gradient and a supported lipid bilayer (SLB) tethered with membrane-bound factors that mimics stromal cell membranes. The chemoattractant channels are separated by hydrogel barriers from SLB in the cell loading channel, which enable precise control of timing and profile of the chemokine gradients applied on cells interacting with SLB. The hydrogel barriers are formed in pillar-free channels through a liquid pinning process, which eliminates complex cleanroom-based fabrications and distortion of chemoattractant gradient by pillars in typical microfluidic hydrogel barrier designs. As a proof-of-concept, we formed an SLB tethered with ICAM-1, and demonstrated its lateral mobility and different migratory behavior of Jurkat T cells on it from those on immobilized ICAM-1, under a gradient of chemokine CXCL12. Our platform can thus be widely used to investigate membrane-bound chemotaxis such as in cancer, immune, and stem cells.
Collapse
Affiliation(s)
- Jia Hao
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA; (J.H.); (W.Z.); (J.M.O.)
| | - Winfield Zhao
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA; (J.H.); (W.Z.); (J.M.O.)
| | - Jeong Min Oh
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA; (J.H.); (W.Z.); (J.M.O.)
| | - Keyue Shen
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA; (J.H.); (W.Z.); (J.M.O.)
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
- USC Stem Cell, University of Southern California, Los Angeles, CA 90033, USA
- Correspondence:
| |
Collapse
|
45
|
Biofabrication of advanced in vitro and ex vivo cancer models for disease modeling and drug screening. FUTURE DRUG DISCOVERY 2021. [DOI: 10.4155/fdd-2020-0034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Bioengineered in vitro models have advanced from 2D cultures and simple 3D cell aggregates to more complex organoids and organ-on-a-chip platforms. This shift has been substantial in cancer research; while simple systems remain in use, multi-tissue type tumor and tissue chips and patient-derived tumor organoids have grown rapidly. These more advanced models offer new tools to cancer researchers based on human tumor physiology and the potential for interactions with nontumor tissue physiology while avoiding critical differences between human and animal biology. In this focused review, the authors discuss the importance of organoid and organ-on-a-chip platforms, with a particular focus on modeling cancer, to highlight oncology-focused in vitro model platform technologies that improve upon the simple 2D cultures and 3D spheroid models of the past.
Collapse
|
46
|
Tan ML, Ling L, Fischbach C. Engineering strategies to capture the biological and biophysical tumor microenvironment in vitro. Adv Drug Deliv Rev 2021; 176:113852. [PMID: 34197895 PMCID: PMC8440401 DOI: 10.1016/j.addr.2021.113852] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 12/11/2022]
Abstract
Despite decades of research and advancements in diagnostic and treatment modalities, cancer remains a major global healthcare challenge. This is due in part to a lack of model systems that allow investigating the mechanisms underlying tumor development, progression, and therapy resistance under relevant conditions in vitro. Tumor cell interactions with their surroundings influence all stages of tumorigenesis and are shaped by both biological and biophysical cues including cell-cell and cell-extracellular matrix (ECM) interactions, tissue architecture and mechanics, and mass transport. Engineered tumor models provide promising platforms to elucidate the individual and combined contributions of these cues to tumor malignancy under controlled and physiologically relevant conditions. This review will summarize current knowledge of the biological and biophysical microenvironmental cues that influence tumor development and progression, present examples of in vitro model systems that are presently used to study these interactions and highlight advancements in tumor engineering approaches to further improve these technologies.
Collapse
Affiliation(s)
- Matthew L Tan
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Lu Ling
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA; Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
47
|
Purkayastha P, Jaiswal MK, Lele TP. Molecular cancer cell responses to solid compressive stress and interstitial fluid pressure. Cytoskeleton (Hoboken) 2021; 78:312-322. [PMID: 34291887 DOI: 10.1002/cm.21680] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 01/19/2023]
Abstract
Alterations to the mechanical properties of the microenvironment are a hallmark of cancer. Elevated mechanical stresses exist in many solid tumors and elicit responses from cancer cells. Uncontrolled growth in confined environments gives rise to elevated solid compressive stress on cancer cells. Recruitment of leaky blood vessels and an absence of functioning lymphatic vessels causes a rise in the interstitial fluid pressure. Here we review the role of the cancer cell cytoskeleton and the nucleus in mediating both the initial and adaptive cancer cell response to these two types of mechanical stresses. We review how these mechanical stresses alter cancer cell functions such as proliferation, apoptosis, and migration.
Collapse
Affiliation(s)
- Purboja Purkayastha
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA
| | - Manish K Jaiswal
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Tanmay P Lele
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA.,Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA.,Department of Translational Medical Sciences, Texas A&M University, Houston, Texas, USA
| |
Collapse
|
48
|
Gilardi M, Bersini S, Valtorta S, Proietto M, Crippa M, Boussommier-Calleja A, Labelle M, Moresco RM, Vanoni M, Kamm RD, Moretti M. The driving role of the Cdk5/Tln1/FAK S732 axis in cancer cell extravasation dissected by human vascularized microfluidic models. Biomaterials 2021; 276:120975. [PMID: 34333365 DOI: 10.1016/j.biomaterials.2021.120975] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 06/04/2021] [Accepted: 06/11/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Understanding the molecular mechanisms of metastatic dissemination, the leading cause of death in cancer patients, is required to develop novel, effective therapies. Extravasation, an essential rate-limiting process in the metastatic cascade, includes three tightly coordinated steps: cancer cell adhesion to the endothelium, trans-endothelial migration, and early invasion into the secondary site. Focal adhesion proteins, including Tln1 and FAK, regulate the cytoskeleton dynamics: dysregulation of these proteins is often associated with metastatic progression and poor prognosis. METHODS Here, we studied the previously unexplored role of these targets in each extravasation step using engineered 3D in vitro models, which recapitulate the physiological vascular niche experienced by cancer cells during hematogenous metastasis. RESULTS Human breast cancer and fibrosarcoma cell lines respond to Cdk5/Tln1/FAK axis perturbation, impairing their metastatic potential. Vascular breaching requires actin polymerization-dependent invadopodia formation. Invadopodia generation requires the structural function of FAK and Tln1 rather than their activation through phosphorylation. Our data support that the inhibition of FAKS732 phosphorylation delocalizes ERK from the nucleus, decreasing ERK phosphorylated form. These findings indicate the critical role of these proteins in driving trans-endothelial migration. In fact, both knock-down experiments and chemical inhibition of FAK dramatically reduces lung colonization in vivo and TEM in microfluidic setting. Altogether, these data indicate that engineered 3D in vitro models coupled to in vivo models, genetic, biochemical, and imaging tools represent a powerful weapon to increase our understanding of metastatic progression. CONCLUSIONS These findings point to the need for further analyses of previously overlooked phosphorylation sites of FAK, such as the serine 732, and foster the development of new effective antimetastatic treatments targeting late events of the metastatic cascade.
Collapse
Affiliation(s)
- Mara Gilardi
- Cell and Tissue Engineering Lab, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy; Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126, Milan, Italy; Institute of Pathology, University Hospital Basel, University of Basel, 4031, Basel, Switzerland.
| | - Simone Bersini
- Cell and Tissue Engineering Lab, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy; Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale, Lugano, Switzerland.
| | - Silvia Valtorta
- Università Degli Studi di Milano-Bicocca, Department of Medicine and Surgery and Tecnomed Foundation, Monza, Italy; Institute of Bioimaging and Molecular Physiology of National Researches Council (IBFM-CNR), Segrate, Italy.
| | - Marco Proietto
- Department of Biology-University of California - San Diego, La Jolla, CA, USA.
| | - Martina Crippa
- Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale, Lugano, Switzerland; Laboratory of Biological Structures Mechanics, Chemistry, Material and Chemical Engineering Department "Giulio Natta", Politecnico di Milano, Milan, Italy.
| | - Alexandra Boussommier-Calleja
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, 02139, MA, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, 02139, MA, USA.
| | - Myriam Labelle
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Rosa Maria Moresco
- Università Degli Studi di Milano-Bicocca, Department of Medicine and Surgery and Tecnomed Foundation, Monza, Italy; Institute of Bioimaging and Molecular Physiology of National Researches Council (IBFM-CNR), Segrate, Italy.
| | - Marco Vanoni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126, Milan, Italy; ISBE.IT/ Centre of Systems Biology, Milano, Italy.
| | - Roger D Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, 02139, MA, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, 02139, MA, USA.
| | - Matteo Moretti
- Cell and Tissue Engineering Lab, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy; Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale, Lugano, Switzerland; Euler Institute, Biomedical Sciences Faculty, Università Della Svizzera Italiana, Lugano, Switzerland.
| |
Collapse
|
49
|
Wang J, Li J, Liu J, Lin M, Mao S, Wang Y, Luo Y. Adsorption Force of Fibronectin: A Balance Regulator to Transmission of Cell Traction Force and Fluid Shear Stress. Biomacromolecules 2021; 22:3264-3273. [PMID: 34225453 DOI: 10.1021/acs.biomac.1c00375] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Osteoblasts actively generate cell traction force (CTF) to sense chemical and mechanical microenvironments. Fluid shear stress (FSS) is a principle mechanical stimulus for bone modeling/remodeling. FSS and CTF share common interconnected elements for force transmission, among which the role of the protein-material interfacial force (Fad) remains unclear. Here, we found that, on the low Fad surface (5.47 ± 1.31 pN/FN), CTF overwhelmed Fad to partially desorb FN, and FSS exacerbated the desorption, resulting in disassembly of the actin cytoskeleton and focal adhesions (FAs) to reduce CTF and establishment of a new mechanical balance at the FN-material interface. Contrarily, on the high Fad surface (27.68 ± 5.24 pN/FN), pure CTF or the combination of CTF and FSS induced no FN desorption, and FSS promoted assembly of actin cytoskeletons and disassembly of FAs, regaining new mechanical balance at the cell-FN interface. These results indicate that Fad is a mechanical regulator for transmission of CTF and FSS, which has never been reported before.
Collapse
Affiliation(s)
- Jinfeng Wang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, and Lab for Smart & Bioinspired Materials, College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Junyao Li
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, and Lab for Smart & Bioinspired Materials, College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Juan Liu
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, and Lab for Smart & Bioinspired Materials, College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Manping Lin
- Key Laboratory of Emergency and Trauma, Ministry of Education, Hainan Medical University, Haikou, 571199, China
| | - Shilong Mao
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, and Lab for Smart & Bioinspired Materials, College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Yuanliang Wang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, and Lab for Smart & Bioinspired Materials, College of Bioengineering, Chongqing University, Chongqing, 400030, China
| | - Yanfeng Luo
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, and Lab for Smart & Bioinspired Materials, College of Bioengineering, Chongqing University, Chongqing, 400030, China
| |
Collapse
|
50
|
Liu X, Fang J, Huang S, Wu X, Xie X, Wang J, Liu F, Zhang M, Peng Z, Hu N. Tumor-on-a-chip: from bioinspired design to biomedical application. MICROSYSTEMS & NANOENGINEERING 2021; 7:50. [PMID: 34567763 PMCID: PMC8433302 DOI: 10.1038/s41378-021-00277-8] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 04/09/2021] [Accepted: 04/15/2021] [Indexed: 05/08/2023]
Abstract
Cancer is one of the leading causes of human death, despite enormous efforts to explore cancer biology and develop anticancer therapies. The main challenges in cancer research are establishing an efficient tumor microenvironment in vitro and exploring efficient means for screening anticancer drugs to reveal the nature of cancer and develop treatments. The tumor microenvironment possesses human-specific biophysical and biochemical factors that are difficult to recapitulate in conventional in vitro planar cell models and in vivo animal models. Therefore, model limitations have hindered the translation of basic research findings to clinical applications. In this review, we introduce the recent progress in tumor-on-a-chip devices for cancer biology research, medicine assessment, and biomedical applications in detail. The emerging tumor-on-a-chip platforms integrating 3D cell culture, microfluidic technology, and tissue engineering have successfully mimicked the pivotal structural and functional characteristics of the in vivo tumor microenvironment. The recent advances in tumor-on-a-chip platforms for cancer biology studies and biomedical applications are detailed and analyzed in this review. This review should be valuable for further understanding the mechanisms of the tumor evolution process, screening anticancer drugs, and developing cancer therapies, and it addresses the challenges and potential opportunities in predicting drug screening and cancer treatment.
Collapse
Affiliation(s)
- Xingxing Liu
- The First Affiliated Hospital of Sun Yat-Sen University, State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, 510006 Guangzhou, China
| | - Jiaru Fang
- The First Affiliated Hospital of Sun Yat-Sen University, State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, 510006 Guangzhou, China
| | - Shuang Huang
- The First Affiliated Hospital of Sun Yat-Sen University, State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, 510006 Guangzhou, China
| | - Xiaoxue Wu
- The First Affiliated Hospital of Sun Yat-Sen University, State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, 510006 Guangzhou, China
| | - Xi Xie
- The First Affiliated Hospital of Sun Yat-Sen University, State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, 510006 Guangzhou, China
| | - Ji Wang
- The First Affiliated Hospital of Sun Yat-Sen University, State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, 510006 Guangzhou, China
| | - Fanmao Liu
- The First Affiliated Hospital of Sun Yat-Sen University, State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, 510006 Guangzhou, China
| | - Meng Zhang
- The First Affiliated Hospital of Sun Yat-Sen University, State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, 510006 Guangzhou, China
| | - Zhenwei Peng
- The First Affiliated Hospital of Sun Yat-Sen University, State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, 510006 Guangzhou, China
| | - Ning Hu
- The First Affiliated Hospital of Sun Yat-Sen University, State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, 510006 Guangzhou, China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, 200050 Shanghai, China
| |
Collapse
|