1
|
Heninger E, Breneman MT, Recchia EE, Kerr SC, Dogru RE, Sharifi MN, LeBeau AM, Kosoff D. Dynamic reciprocal interactions between activated T cells and tumor associated macrophages drive macrophage reprogramming and proinflammatory T cell migration within prostate tumor models. Sci Rep 2024; 14:24230. [PMID: 39414902 PMCID: PMC11484957 DOI: 10.1038/s41598-024-75265-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024] Open
Abstract
Tumor-associated macrophages (TAMs) have been implicated as a tumor microenvironment (TME) cell population, which may be playing a vital role in the inhibition of effective T cell responses in the prostate TME. In this manuscript, we leverage a novel microscale cell culture platform, known as Stacks, to investigate mono-, co-, and tri-culture TME models comprised of prostate tumor cell lines, primary macrophages, and autologous T cells from patients with prostate cancer. Through multiplexed analysis of these multi-cellular prostate tumor models, we capture a dynamic interaction between primary TAMs and activated T cells that resulted in reciprocal proinflammatory activation of both cell populations upon interaction. These findings suggest that activated T cells are capable of reprogramming immunosuppressive TAMs in the context of prostate tumor models and that TAM reprogramming may play a key supportive role in restoring proinflammatory T cell tumor responses in the prostate TME.
Collapse
Affiliation(s)
- Erika Heninger
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | | | | | - Sheena Catherine Kerr
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Reyna Elvan Dogru
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Aaron Matthew LeBeau
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - David Kosoff
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.
- William S Middleton Memorial Veteran's Hospital, Madison, WI, USA.
- Department of Medicine, Carbone Cancer Center, University of Wisconsin Madison, 1111 Highland Avenue, WIMR 7105, Madison, WI, USA.
| |
Collapse
|
2
|
Haack AJ, Brown LG, Goldstein AJ, Mulimani P, Berthier J, Viswanathan AR, Kopyeva I, Whitten JM, Lin A, Nguyen SH, Leahy TP, Bouker EE, Padgett RM, Mazzawi NA, Tokihiro JC, Bretherton RC, Wu A, Tapscott SJ, DeForest CA, Popowics TE, Berthier E, Sniadecki NJ, Theberge AB. Suspended Tissue Open Microfluidic Patterning (STOMP). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.04.616662. [PMID: 39416011 PMCID: PMC11482760 DOI: 10.1101/2024.10.04.616662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Cell-laden hydrogel constructs suspended between pillars are powerful tools for modeling tissue structure and physiology, though current fabrication techniques often limit them to uniform compositions. In contrast, tissues are complex in nature with spatial arrangements of cell types and extracellular matrices. Thus, we present Suspended Tissue Open Microfluidic Patterning (STOMP), which utilizes a removable, open microfluidic patterning channel to pattern multiple spatial regions across a single suspended tissue. The STOMP platform contains capillary pinning features along the open channel that controls the fluid front, allowing multiple cell and extracellular matrix precursors to be pipetted into one tissue. We have used this technique to pattern suspended tissues with multiple regional components using a variety of native and synthetic extracellular matrices, including fibrin, collagen, and poly(ethylene glycol). Here, we demonstrate that STOMP models a region of fibrosis in a functional heart tissue and a bone-ligament junction in periodontal tissues. Additionally, the STOMP platform can be customized to allow patterning of suspended cores and more spatial configurations, enhancing its utility in complex tissue modeling. STOMP is a versatile technique for generating suspended tissue models with increased control over cell and hydrogel composition to model interfacial tissue regions in a suspended tissue.
Collapse
Affiliation(s)
- Amanda J. Haack
- Department of Chemistry, University of Washington, Seattle, WA, 98195 USA
- Medical Scientist Training Program, University of Washington School of Medicine, Seattle, WA, 98195 USA
| | - Lauren G. Brown
- Department of Chemistry, University of Washington, Seattle, WA, 98195 USA
| | - Alex J. Goldstein
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98109 USA
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195 USA
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, 98195 USA
| | - Priti Mulimani
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98109 USA
- Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA, 98195 USA
| | - Jean Berthier
- Department of Chemistry, University of Washington, Seattle, WA, 98195 USA
| | | | - Irina Kopyeva
- Department of Bioengineering, University of Washington, Seattle, WA, 98195 USA
| | - Jamison M. Whitten
- Department of Chemistry, University of Washington, Seattle, WA, 98195 USA
| | - Ariel Lin
- Department of Chemistry, University of Washington, Seattle, WA, 98195 USA
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA, 98109 USA
| | - Serena H. Nguyen
- Department of Chemistry, University of Washington, Seattle, WA, 98195 USA
| | - Thomas P. Leahy
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98109 USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, 98195 USA
| | - Ella E. Bouker
- Department of Chemistry, University of Washington, Seattle, WA, 98195 USA
| | - Ruby M. Padgett
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98109 USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, 98195 USA
| | - Natalie A. Mazzawi
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98109 USA
- Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA, 98195 USA
- Department of Microbiology, University of Washington, Seattle, WA, 98195 USA
| | - Jodie C. Tokihiro
- Department of Chemistry, University of Washington, Seattle, WA, 98195 USA
| | - Ross C. Bretherton
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98109 USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195 USA
| | - Aaliyah Wu
- Department of Chemistry, University of Washington, Seattle, WA, 98195 USA
| | - Stephen J. Tapscott
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Department of Neurology, University of Washington, Seattle WA 98195, USA
| | - Cole A. DeForest
- Department of Chemistry, University of Washington, Seattle, WA, 98195 USA
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98109 USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195 USA
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA, 98109 USA
- Department of Chemical Engineering, University of Washington, Seattle, WA, 98195 USA
- Institute for Protein Design, University of Washington, Seattle, WA, 98195 USA
| | - Tracy E. Popowics
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98109 USA
- Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA, 98195 USA
| | - Erwin Berthier
- Department of Chemistry, University of Washington, Seattle, WA, 98195 USA
| | - Nathan J. Sniadecki
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98109 USA
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, 98195 USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195 USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, 98195 USA
| | - Ashleigh B. Theberge
- Department of Chemistry, University of Washington, Seattle, WA, 98195 USA
- Department of Urology, University of Washington School of Medicine, Seattle, WA, 98195 USA
| |
Collapse
|
3
|
Liu X, Gao M, Li B, Liu R, Chong Z, Gu Z, Zhou K. Bioinspired Capillary Transistors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310797. [PMID: 39139014 DOI: 10.1002/adma.202310797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 05/29/2024] [Indexed: 08/15/2024]
Abstract
Inspired by the unidirectional liquid spreading on Nepenthes peristome, Araucaria leaf, butterfly wings, etc., various microfluidic devices have been developed for water collection, irrigation, physical/chemical reaction, and oil-water separation. Despite extensive progress, most natural and artificial structures fail to enhance the Laplace pressure difference or capillary force, thus suffering from a low unidirectional capillary height (<30 mm). In this work, asymmetric re-entrant structures with long overhangs and connected forward/lateral microchannels are fabricated by 3D printing, resulting in a significantly increased unidirectional capillary height of 102.3 mm for water, which approximately corresponds to the theoretical limit. The overhangs can partially overlap the forward microchannels of the front structures without direct contact, thus enhancing the Laplace pressure difference and capillary force simultaneously. Based on asymmetric and symmetric re-entrant structures, capillary transistors are proposed and realized to programmably adjust the capillary direction, height, and width, which are envisioned to function as switches/valves and amplifiers/attenuators for highly efficient liquid patterning, desalination, and biochemical microreaction in 3D space.
Collapse
Affiliation(s)
- Xiaojiang Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- HP-NTU Digital Manufacturing Corporate Lab, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| | - Ming Gao
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| | - Boyuan Li
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| | - Ruoyu Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Zhejun Chong
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Zhongze Gu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Kun Zhou
- HP-NTU Digital Manufacturing Corporate Lab, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| |
Collapse
|
4
|
Lin L, Bao Y. Development and validation of machine learning models for diagnosis and prognosis of lung adenocarcinoma, and immune infiltration analysis. Sci Rep 2024; 14:22081. [PMID: 39333719 PMCID: PMC11437281 DOI: 10.1038/s41598-024-73498-2] [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: 04/17/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024] Open
Abstract
The aim of our study was to develop robust diagnostic and prognostic models for lung adenocarcinoma (LUAD) using machine learning (ML) techniques, focusing on early immune infiltration. Feature selection was performed on The Cancer Genome Atlas (TCGA) data using least absolute shrinkage and selection Operator (LASSO), random forest (RF), and support vector machine (SVM) algorithms. Six ML algorithms were employed to construct the diagnostic models, which were evaluated through receiver operating characteristic (ROC) curves, precision-recall curves (PRC), and classification error (CE), and validated on the GSE7670 dataset. Additionally, a lasso cox prognostic model was built on the TCGA-LUAD dataset and externally validated using independent Gene Expression Omnibus datasets (GSE30219, GSE31210, GSE50081, and GSE37745). Single-sample gene set enrichment analysis (ssGSEA) was performed to assess immune cell infiltration in stage I LUAD samples, revealing significant differences in immune cell types. These findings demonstrate a positive correlation between immune infiltration in stage I LUAD and Th2 cells, Tcm cells, and T helper cells, while a negative correlation was observed with Macrophages, Eosinophils, and Tem cells. These insights provide novel perspectives for clinical diagnosis and treatment of LUAD.
Collapse
Affiliation(s)
- Lin Lin
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, No. 148, Health Care Road, Nangang District, Harbin, 150086, Heilongjiang, People's Republic of China
| | - Yongxia Bao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, No. 148, Health Care Road, Nangang District, Harbin, 150086, Heilongjiang, People's Republic of China.
| |
Collapse
|
5
|
Gaebler D, Hachey SJ, Hughes CCW. Improving tumor microenvironment assessment in chip systems through next-generation technology integration. Front Bioeng Biotechnol 2024; 12:1462293. [PMID: 39386043 PMCID: PMC11461320 DOI: 10.3389/fbioe.2024.1462293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/10/2024] [Indexed: 10/12/2024] Open
Abstract
The tumor microenvironment (TME) comprises a diverse array of cells, both cancerous and non-cancerous, including stromal cells and immune cells. Complex interactions among these cells play a central role in driving cancer progression, impacting critical aspects such as tumor initiation, growth, invasion, response to therapy, and the development of drug resistance. While targeting the TME has emerged as a promising therapeutic strategy, there is a critical need for innovative approaches that accurately replicate its complex cellular and non-cellular interactions; the goal being to develop targeted, personalized therapies that can effectively elicit anti-cancer responses in patients. Microfluidic systems present notable advantages over conventional in vitro 2D co-culture models and in vivo animal models, as they more accurately mimic crucial features of the TME and enable precise, controlled examination of the dynamic interactions among multiple human cell types at any time point. Combining these models with next-generation technologies, such as bioprinting, single cell sequencing and real-time biosensing, is a crucial next step in the advancement of microfluidic models. This review aims to emphasize the importance of this integrated approach to further our understanding of the TME by showcasing current microfluidic model systems that integrate next-generation technologies to dissect cellular intra-tumoral interactions across different tumor types. Carefully unraveling the complexity of the TME by leveraging next generation technologies will be pivotal for developing targeted therapies that can effectively enhance robust anti-tumoral responses in patients and address the limitations of current treatment modalities.
Collapse
Affiliation(s)
- Daniela Gaebler
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Stephanie J. Hachey
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Christopher C. W. Hughes
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
- Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
6
|
Ignes-Romeu A, Weppner HK, Kaur T, Singh M, Hind LE. THP-1 Macrophages Limit Neutrophil Transendothelial Migration in a Model Infection. Cell Mol Bioeng 2024; 17:279-293. [PMID: 39372553 PMCID: PMC11450111 DOI: 10.1007/s12195-024-00813-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 07/09/2024] [Indexed: 10/08/2024] Open
Abstract
Introduction Dysregulated neutrophil function plays a significant role in the pathology of infections, cancer, cardiovascular diseases, and autoimmune disorders. Neutrophil activity is influenced by various cell populations, including macrophages, which are crucial regulators. However, the exact role of human macrophages in controlling neutrophil function remains unclear due to a scarcity of studies utilizing human cells in physiologically relevant models. Methods We adapted our "Infection-on-a-Chip" microfluidic device to incorporate macrophages within the collagen extracellular matrix, allowing for the study of interactions between human neutrophils and macrophages in a context that mimics in vivo conditions. The integration of THP-1 macrophages was optimized and their effect on the endothelial lumen was characterized, focusing on permeability and structural integrity. The device was then employed to examine the influence of macrophages on neutrophil response to infection with the bacterial pathogen Pseudomonas aeruginosa. Results Integration of THP-1 macrophages into the microfluidic device was successfully optimized, showing no increase in endothelial permeability or structural damage. The presence of macrophages was found to significantly reduce neutrophil transendothelial migration in response to Pseudomonas aeruginosa infection. Conclusions Our findings highlight the regulatory role of macrophages in modulating neutrophil responses, suggesting potential therapeutic targets to control neutrophil function in various diseases. The modified microfluidic platform offers a valuable tool for mechanistic studies into macrophage-neutrophil interactions in disease contexts. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-024-00813-2.
Collapse
Affiliation(s)
- Aitana Ignes-Romeu
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO 80303 USA
| | - Hannah K. Weppner
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO 80303 USA
| | - Tanisha Kaur
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO 80303 USA
| | - Maya Singh
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO 80303 USA
- Present Address: Department of Bioengineering, University of Washington, Seattle, WA 98195 USA
| | - Laurel E. Hind
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO 80303 USA
| |
Collapse
|
7
|
Nebuloni F, Deroy C, Cook PR, Walsh EJ. Stable diffusion gradients in microfluidic conduits bounded by fluid walls. MICROSYSTEMS & NANOENGINEERING 2024; 10:79. [PMID: 38911344 PMCID: PMC11189932 DOI: 10.1038/s41378-024-00698-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 04/02/2024] [Indexed: 06/25/2024]
Abstract
Assays mimicking in vitro the concentration gradients triggering biological responses like those involved in fighting infections and blood clotting are essential for biomedical research. Microfluidic assays prove especially attractive as they allow precise control of gradient shape allied to a reduction in scale. Conventional microfluidic devices are fabricated using solid plastics that prevent direct access to responding cells. Fluid-walled microfluidics allows the manufacture of circuits on standard Petri dishes in seconds, coupled to simple operating methods; cell-culture medium sitting in a standard dish is confined to circuits by fluid walls made of an immiscible fluorocarbon. We develop and experimentally validate an analytical model of diffusion between two or more aqueous streams flowing at different rates into a fluid-walled conduit with the cross-section of a circular segment. Unlike solid walls, fluid walls morph during flows as pressures fall, with wall shape changing down the conduit. The model is validated experimentally for Fourier numbers < 0.1 using fluorescein diffusing between laminar streams. It enables a priori prediction of concentration gradients throughout a conduit, so allowing rapid circuit design as well as providing bio-scientists with an accurate way of predicting local concentrations of bioactive molecules around responsive and non-responsive cells.
Collapse
Affiliation(s)
- Federico Nebuloni
- Department of Engineering Science, Osney Thermo-Fluids Institute, University of Oxford, Oxford, OX2 0ES UK
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE UK
| | - Cyril Deroy
- Department of Engineering Science, Osney Thermo-Fluids Institute, University of Oxford, Oxford, OX2 0ES UK
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE UK
| | - Peter R. Cook
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE UK
| | - Edmond J. Walsh
- Department of Engineering Science, Osney Thermo-Fluids Institute, University of Oxford, Oxford, OX2 0ES UK
| |
Collapse
|
8
|
Cadavid JL, Li NT, McGuigan AP. Bridging systems biology and tissue engineering: Unleashing the full potential of complex 3D in vitro tissue models of disease. BIOPHYSICS REVIEWS 2024; 5:021301. [PMID: 38617201 PMCID: PMC11008916 DOI: 10.1063/5.0179125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 03/12/2024] [Indexed: 04/16/2024]
Abstract
Rapid advances in tissue engineering have resulted in more complex and physiologically relevant 3D in vitro tissue models with applications in fundamental biology and therapeutic development. However, the complexity provided by these models is often not leveraged fully due to the reductionist methods used to analyze them. Computational and mathematical models developed in the field of systems biology can address this issue. Yet, traditional systems biology has been mostly applied to simpler in vitro models with little physiological relevance and limited cellular complexity. Therefore, integrating these two inherently interdisciplinary fields can result in new insights and move both disciplines forward. In this review, we provide a systematic overview of how systems biology has been integrated with 3D in vitro tissue models and discuss key application areas where the synergies between both fields have led to important advances with potential translational impact. We then outline key directions for future research and discuss a framework for further integration between fields.
Collapse
|
9
|
Gaebler D, Hachey SJ, Hughes CCW. Microphysiological systems as models for immunologically 'cold' tumors. Front Cell Dev Biol 2024; 12:1389012. [PMID: 38711620 PMCID: PMC11070549 DOI: 10.3389/fcell.2024.1389012] [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: 02/20/2024] [Accepted: 03/25/2024] [Indexed: 05/08/2024] Open
Abstract
The tumor microenvironment (TME) is a diverse milieu of cells including cancerous and non-cancerous cells such as fibroblasts, pericytes, endothelial cells and immune cells. The intricate cellular interactions within the TME hold a central role in shaping the dynamics of cancer progression, influencing pivotal aspects such as tumor initiation, growth, invasion, response to therapeutic interventions, and the emergence of drug resistance. In immunologically 'cold' tumors, the TME is marked by a scarcity of infiltrating immune cells, limited antigen presentation in the absence of potent immune-stimulating signals, and an abundance of immunosuppressive factors. While strategies targeting the TME as a therapeutic avenue in 'cold' tumors have emerged, there is a pressing need for novel approaches that faithfully replicate the complex cellular and non-cellular interactions in order to develop targeted therapies that can effectively stimulate immune responses and improve therapeutic outcomes in patients. Microfluidic devices offer distinct advantages over traditional in vitro 3D co-culture models and in vivo animal models, as they better recapitulate key characteristics of the TME and allow for precise, controlled insights into the dynamic interplay between various immune, stromal and cancerous cell types at any timepoint. This review aims to underscore the pivotal role of microfluidic systems in advancing our understanding of the TME and presents current microfluidic model systems that aim to dissect tumor-stromal, tumor-immune and immune-stromal cellular interactions in various 'cold' tumors. Understanding the intricacies of the TME in 'cold' tumors is crucial for devising effective targeted therapies to reinvigorate immune responses and overcome the challenges of current immunotherapy approaches.
Collapse
Affiliation(s)
- Daniela Gaebler
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Stephanie J. Hachey
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Christopher C. W. Hughes
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
- Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
10
|
Sassi A, You L. Microfluidics-Based Technologies for the Assessment of Castration-Resistant Prostate Cancer. Cells 2024; 13:575. [PMID: 38607014 PMCID: PMC11011521 DOI: 10.3390/cells13070575] [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: 02/22/2024] [Revised: 03/22/2024] [Accepted: 03/23/2024] [Indexed: 04/13/2024] Open
Abstract
Castration-resistant prostate cancer remains a significant clinical challenge, wherein patients display no response to existing hormone therapies. The standard of care often includes aggressive treatment options using chemotherapy, radiation therapy and various drugs to curb the growth of additional metastases. As such, there is a dire need for the development of innovative technologies for both its diagnosis and its management. Traditionally, scientific exploration of prostate cancer and its treatment options has been heavily reliant on animal models and two-dimensional (2D) in vitro technologies. However, both laboratory tools often fail to recapitulate the dynamic tumor microenvironment, which can lead to discrepancies in drug efficacy and side effects in a clinical setting. In light of the limitations of traditional animal models and 2D in vitro technologies, the emergence of microfluidics as a tool for prostate cancer research shows tremendous promise. Namely, microfluidics-based technologies have emerged as powerful tools for assessing prostate cancer cells, isolating circulating tumor cells, and examining their behaviour using tumor-on-a-chip models. As such, this review aims to highlight recent advancements in microfluidics-based technologies for the assessment of castration-resistant prostate cancer and its potential to advance current understanding and to improve therapeutic outcomes.
Collapse
Affiliation(s)
- Amel Sassi
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada;
| | - Lidan You
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada;
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Department of Mechanical and Materials Engineering, Queen’s University, Kingston, ON K7L 2V9, Canada
| |
Collapse
|
11
|
Wu Z, Huang D, Wang J, Zhao Y, Sun W, Shen X. Engineering Heterogeneous Tumor Models for Biomedical Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304160. [PMID: 37946674 PMCID: PMC10767453 DOI: 10.1002/advs.202304160] [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: 06/22/2023] [Revised: 09/16/2023] [Indexed: 11/12/2023]
Abstract
Tumor tissue engineering holds great promise for replicating the physiological and behavioral characteristics of tumors in vitro. Advances in this field have led to new opportunities for studying the tumor microenvironment and exploring potential anti-cancer therapeutics. However, the main obstacle to the widespread adoption of tumor models is the poor understanding and insufficient reconstruction of tumor heterogeneity. In this review, the current progress of engineering heterogeneous tumor models is discussed. First, the major components of tumor heterogeneity are summarized, which encompasses various signaling pathways, cell proliferations, and spatial configurations. Then, contemporary approaches are elucidated in tumor engineering that are guided by fundamental principles of tumor biology, and the potential of a bottom-up approach in tumor engineering is highlighted. Additionally, the characterization approaches and biomedical applications of tumor models are discussed, emphasizing the significant role of engineered tumor models in scientific research and clinical trials. Lastly, the challenges of heterogeneous tumor models in promoting oncology research and tumor therapy are described and key directions for future research are provided.
Collapse
Affiliation(s)
- Zhuhao Wu
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Danqing Huang
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Jinglin Wang
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Yuanjin Zhao
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalWenzhou Medical UniversityWenzhou325035China
| | - Weijian Sun
- Department of Gastrointestinal SurgeryThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Xian Shen
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalWenzhou Medical UniversityWenzhou325035China
| |
Collapse
|
12
|
Kyriakopoulou K, Koutsakis C, Piperigkou Z, Karamanos NK. Recreating the extracellular matrix: novel 3D cell culture platforms in cancer research. FEBS J 2023; 290:5238-5247. [PMID: 36929947 DOI: 10.1111/febs.16778] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/23/2023] [Accepted: 03/15/2023] [Indexed: 03/18/2023]
Abstract
Cancer initiation and progression heavily rely on microenvironmental cues derived from various components of the niche including the extracellular matrix (ECM). ECM is a complex macromolecular network that governs cell functionality. Although the two-dimensional (2D) cell culture systems provide useful information at the molecular level and preclinical testing, they could not accurately represent the in vivo matrix microenvironmental architecture. Hence, it is no surprise that researchers in the last decade have focussed their efforts on establishing novel advanced in vitro culture models that mimic tumour and tissue-specific niches and interactions. These numerous three-dimensional (3D) culture systems that are now widely available, as well as those still under development, grant researchers with new, improved tools to study cancer progression and to explore innovative therapeutic options. Herein, we report on the emerging methods and cutting-edge technologies in 3D cell culture platforms and discuss their potential use in unveiling tumour microenvironmental cues, drug screening and personalized treatment.
Collapse
Affiliation(s)
- Konstantina Kyriakopoulou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Christos Koutsakis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| |
Collapse
|
13
|
Luo Q, Shang K, Zhu J, Wu Z, Cao T, Ahmed AAQ, Huang C, Xiao L. Biomimetic cell culture for cell adhesive propagation for tissue engineering strategies. MATERIALS HORIZONS 2023; 10:4662-4685. [PMID: 37705440 DOI: 10.1039/d3mh00849e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Biomimetic cell culture, which involves creating a biomimetic microenvironment for cells in vitro by engineering approaches, has aroused increasing interest given that it maintains the normal cellular phenotype, genotype and functions displayed in vivo. Therefore, it can provide a more precise platform for disease modelling, drug development and regenerative medicine than the conventional plate cell culture. In this review, initially, we discuss the principle of biomimetic cell culture in terms of the spatial microenvironment, chemical microenvironment, and physical microenvironment. Then, the main strategies of biomimetic cell culture and their state-of-the-art progress are summarized. To create a biomimetic microenvironment for cells, a variety of strategies has been developed, ranging from conventional scaffold strategies, such as macroscopic scaffolds, microcarriers, and microgels, to emerging scaffold-free strategies, such as spheroids, organoids, and assembloids, to simulate the native cellular microenvironment. Recently, 3D bioprinting and microfluidic chip technology have been applied as integrative platforms to obtain more complex biomimetic structures. Finally, the challenges in this area are discussed and future directions are discussed to shed some light on the community.
Collapse
Affiliation(s)
- Qiuchen Luo
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China.
| | - Keyuan Shang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China.
| | - Jing Zhu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China.
| | - Zhaoying Wu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China.
| | - Tiefeng Cao
- Department of Gynaecology, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510070, China
| | - Abeer Ahmed Qaed Ahmed
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, 27100 Pavia, Italy
| | - Chixiang Huang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China.
| | - Lin Xiao
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China.
| |
Collapse
|
14
|
Liu X, Li B, Gu Z, Zhou K. 4D Printing of Butterfly Scale-Inspired Structures for Wide-Angle Directional Liquid Transport. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2207640. [PMID: 37078893 DOI: 10.1002/smll.202207640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Indexed: 05/03/2023]
Abstract
Unidirectional liquid transport has been extensively explored for water/fog harvesting, electrochemical sensing, and desalination. However, current research mainly focuses on linear liquid transport (transport angle α = 0°), which exhibits hindered lateral liquid spreading and low unidirectional transport efficiency. Inspired by the wide-angle (0° < α < 180°) liquid transport on butterfly wings, this work successfully achieves linear (α = 0°), wide-angle, and even ultra-wide-angle (α = 180°) liquid transport by four-dimensional (4D) printing of butterfly scale-inspired re-entrant structures. These asymmetric re-entrant structures can achieve unidirectional liquid transport, and their layout can control the Laplace pressure in the forward (structure-tilting) and lateral directions to adjust the transport angle. Specifically, high transport efficiency and programmable forward/lateral transport paths are simultaneously achieved by the ultra-wide-angle transport, where liquid fills the lateral path before being transported forward. Moreover, the ultra-wide-angle transport is also validated in 3D space, which provides an innovative platform for advanced biochemical microreaction, large-area evaporation, and self-propelled oil-water separation.
Collapse
Affiliation(s)
- Xiaojiang Liu
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore
- HP-NTU Digital Manufacturing Corporate Lab, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| | - Boyuan Li
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Kun Zhou
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore
- HP-NTU Digital Manufacturing Corporate Lab, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| |
Collapse
|
15
|
Zeng Y, Khor JW, van Neel TL, Tu WC, Berthier J, Thongpang S, Berthier E, Theberge AB. Miniaturizing chemistry and biology using droplets in open systems. Nat Rev Chem 2023; 7:439-455. [PMID: 37117816 PMCID: PMC10107581 DOI: 10.1038/s41570-023-00483-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2023] [Indexed: 04/30/2023]
Abstract
Open droplet microfluidic systems manipulate droplets on the picolitre-to-microlitre scale in an open environment. They combine the compartmentalization and control offered by traditional droplet-based microfluidics with the accessibility and ease-of-use of open microfluidics, bringing unique advantages to applications such as combinatorial reactions, droplet analysis and cell culture. Open systems provide direct access to droplets and allow on-demand droplet manipulation within the system without needing pumps or tubes, which makes the systems accessible to biologists without sophisticated setups. Furthermore, these systems can be produced with simple manufacturing and assembly steps that allow for manufacturing at scale and the translation of the method into clinical research. This Review introduces the different types of open droplet microfluidic system, presents the physical concepts leveraged by these systems and highlights key applications.
Collapse
Affiliation(s)
- Yuting Zeng
- Department of Chemistry, University of Washington, Seattle, WA, USA
| | - Jian Wei Khor
- Department of Chemistry, University of Washington, Seattle, WA, USA
| | - Tammi L van Neel
- Department of Chemistry, University of Washington, Seattle, WA, USA
| | - Wan-Chen Tu
- Department of Chemistry, University of Washington, Seattle, WA, USA
| | - Jean Berthier
- Department of Chemistry, University of Washington, Seattle, WA, USA
| | - Sanitta Thongpang
- Department of Chemistry, University of Washington, Seattle, WA, USA
- Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakorn Pathom, Thailand
| | - Erwin Berthier
- Department of Chemistry, University of Washington, Seattle, WA, USA.
| | - Ashleigh B Theberge
- Department of Chemistry, University of Washington, Seattle, WA, USA.
- Department of Urology, School of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
16
|
Nunn LR, Juang TD, Beebe DJ, Wheeler NJ, Zamanian M. A high-throughput nematode sensory assay reveals an inhibitory effect of ivermectin on parasite gustation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.25.538347. [PMID: 37163046 PMCID: PMC10168391 DOI: 10.1101/2023.04.25.538347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Sensory pathways first elucidated in Caenorhabditis elegans are conserved across free-living and parasitic nematodes, even though each species responds to a diverse array of compounds. Most nematode sensory assays are performed by tallying observations of worm behavior on two-dimensional planes using agarose plates. These assays have been successful in the study of volatile sensation but are poorly suited for investigation of water-soluble gustation or parasitic nematodes without a free-living stage. In contrast, gustatory assays tend to be tedious, often limited to the manipulation of a single individual at a time. We have designed a nematode sensory assay using a microfluidics device that allows for the study of gustation in a 96-well, three-dimensional environment. This device is suited for free-living worms and parasitic worms that spend their lives in an aqueous environment, and we have used it to show that ivermectin inhibits the gustatory ability of vector-borne parasitic nematodes.
Collapse
Affiliation(s)
- Leonardo R. Nunn
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI USA
| | - Terry D. Juang
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI USA
| | - David J. Beebe
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI USA
| | - Nicolas J. Wheeler
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI USA
- Department of Biology, University of Wisconsin-Eau Claire, Eau Claire, WI USA
| | - Mostafa Zamanian
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI USA
| |
Collapse
|
17
|
Ye WQ, Liu XP, Ma RF, Yang CG, Xu ZR. Open-channel microfluidic chip based on shape memory polymer for controllable liquid transport. LAB ON A CHIP 2023; 23:2068-2074. [PMID: 36928455 DOI: 10.1039/d3lc00027c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Open microfluidics has attracted increasing attention over the last decade because of its flexibility and simplicity with respect to cell culture and clinical diagnosis. However, traditional valves and pumps are difficult to integrate on open-channel microfluidic chips, in which a liquid is usually driven by capillary forces. Poor fluid control performance is a common drawback of open microfluidics. Herein, we proposed a method for controlling the liquid flow in open channels by controlling the continuous Laplace pressure induced by the deformation of the shape memory microstructures. The uniformly arranged cuboidal microcolumns in the open channels have magnetic/light dual responses, and the bending angle of the microcolumns can be controlled by adjusting Laplace pressure using near-infrared laser irradiation in a magnetic field. Laplace pressure and capillary force drove the liquid flow together, and the controllable fluid transport was realized by adjusting the hydrophilicity of the channel surface and the bending angle of the microcolumns. We demonstrated the controllability of the flow rate and the directional transport of water along a preset path. In addition, the start and stop of water transport were realized via local hydrophobic modification. The proposed strategy improves poor fluid control in traditional open systems and makes fluid flow highly controllable. We tried to extract and detect rhodamine B in tiny droplets on the open microfluidic chip, demonstrating the advantages of the proposed strategy in the separation and analysis of tiny samples.
Collapse
Affiliation(s)
- Wen-Qi Ye
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, China.
| | - Xiao-Peng Liu
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, China.
| | - Ruo-Fei Ma
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, China.
| | - Chun-Guang Yang
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, China.
| | - Zhang-Run Xu
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, China.
| |
Collapse
|
18
|
Nunn LR, Juang TD, Beebe DJ, Wheeler NJ, Zamanian M. A high-throughput sensory assay for parasitic and free-living nematodes. Integr Biol (Camb) 2023; 15:zyad010. [PMID: 37555835 PMCID: PMC10752570 DOI: 10.1093/intbio/zyad010] [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: 05/18/2023] [Revised: 07/12/2023] [Accepted: 07/14/2023] [Indexed: 08/10/2023]
Abstract
Sensory pathways first elucidated in Caenorhabditis elegans are conserved across free-living and parasitic nematodes, even though each species responds to a diverse array of compounds. Most nematode sensory assays are performed by tallying observations of worm behavior on two-dimensional planes using agarose plates. These assays have been successful in the study of volatile sensation but are poorly suited for investigation of water-soluble gustation or parasitic nematodes without a free-living stage. In contrast, gustatory assays tend to be tedious, often limited to the manipulation of a single individual at a time. We have designed a nematode sensory assay using a microfluidics device that allows for the study of gustation in a 96-well, three-dimensional environment. This device is suited for free-living worms and parasitic worms that spend their lives in an aqueous environment, and we have used it to show that ivermectin inhibits the gustatory ability of vector-borne parasitic nematodes. Insight box Nematodes are powerful model organisms for understanding the sensory biology of multicellular eukaryotes, and many parasitic species cause disease in humans. Simple sensory assays performed on agarose plates have been the bedrock for establishing the neuronal, genetic, and developmental foundations for many sensory modalities in nematodes. However, these classical assays are poorly suited for translational movement of many parasitic nematodes and the sensation of water-soluble molecules (gustation). We have designed a device for high-throughput nematode sensory assays in a gel matrix. This 'gustatory microplate' is amenable to several species and reveals novel responses by free-living and parasitic nematodes to cues and drugs.
Collapse
Affiliation(s)
- Leonardo R. Nunn
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI USA
| | - Terry D. Juang
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI USA
| | - David J. Beebe
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI USA
| | - Nicolas J. Wheeler
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI USA
- Department of Biology, University of Wisconsin-Eau Claire, Eau Claire, WI USA
| | - Mostafa Zamanian
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI USA
| |
Collapse
|
19
|
Manduca N, Maccafeo E, De Maria R, Sistigu A, Musella M. 3D cancer models: One step closer to in vitro human studies. Front Immunol 2023; 14:1175503. [PMID: 37114038 PMCID: PMC10126361 DOI: 10.3389/fimmu.2023.1175503] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/23/2023] [Indexed: 04/29/2023] Open
Abstract
Cancer immunotherapy is the great breakthrough in cancer treatment as it displayed prolonged progression-free survival over conventional therapies, yet, to date, in only a minority of patients. In order to broad cancer immunotherapy clinical applicability some roadblocks need to be overcome, first among all the lack of preclinical models that faithfully depict the local tumor microenvironment (TME), which is known to dramatically affect disease onset, progression and response to therapy. In this review, we provide the reader with a detailed overview of current 3D models developed to mimick the complexity and the dynamics of the TME, with a focus on understanding why the TME is a major target in anticancer therapy. We highlight the advantages and translational potentials of tumor spheroids, organoids and immune Tumor-on-a-Chip models in disease modeling and therapeutic response, while outlining pending challenges and limitations. Thinking forward, we focus on the possibility to integrate the know-hows of micro-engineers, cancer immunologists, pharmaceutical researchers and bioinformaticians to meet the needs of cancer researchers and clinicians interested in using these platforms with high fidelity for patient-tailored disease modeling and drug discovery.
Collapse
Affiliation(s)
- Nicoletta Manduca
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ester Maccafeo
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ruggero De Maria
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario ‘A. Gemelli’ - Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Antonella Sistigu
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Martina Musella
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
20
|
Hou C, Gu Y, Yuan W, Zhang W, Xiu X, Lin J, Gao Y, Liu P, Chen X, Song L. Application of microfluidic chips in the simulation of the urinary system microenvironment. Mater Today Bio 2023; 19:100553. [PMID: 36747584 PMCID: PMC9898763 DOI: 10.1016/j.mtbio.2023.100553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/01/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023] Open
Abstract
The urinary system, comprising the kidneys, ureters, bladder, and urethra, has a unique mechanical and fluid microenvironment, which is essential to the urinary system growth and development. Microfluidic models, based on micromachining and tissue engineering technology, can integrate pathophysiological characteristics, maintain cell-cell and cell-extracellular matrix interactions, and accurately simulate the vital characteristics of human tissue microenvironments. Additionally, these models facilitate improved visualization and integration and meet the requirements of the laminar flow environment of the urinary system. However, several challenges continue to impede the development of a tissue microenvironment with controllable conditions closely resemble physiological conditions. In this review, we describe the biochemical and physical microenvironment of the urinary system and explore the feasibility of microfluidic technology in simulating the urinary microenvironment and pathophysiological characteristics in vitro. Moreover, we summarize the current research progress on adapting microfluidic chips for constructing the urinary microenvironment. Finally, we discuss the current challenges and suggest directions for future development and application of microfluidic technology in constructing the urinary microenvironment in vitro.
Collapse
Affiliation(s)
- Changhao Hou
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, China
| | - Yubo Gu
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, China
| | - Wei Yuan
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, China
| | - Wukai Zhang
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xianjie Xiu
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, China
| | - Jiahao Lin
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, China
| | - Yue Gao
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Peichuan Liu
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiang Chen
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Lujie Song
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, China
| |
Collapse
|
21
|
Ferrari D, Sengupta A, Heo L, Pethö L, Michler J, Geiser T, de Jesus Perez VA, Kuebler WM, Zeinali S, Guenat OT. Effects of biomechanical and biochemical stimuli on angio- and vasculogenesis in a complex microvasculature-on-chip. iScience 2023; 26:106198. [PMID: 36879808 PMCID: PMC9985038 DOI: 10.1016/j.isci.2023.106198] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/31/2022] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
The endothelium of blood vessels is a vital organ that reacts differently to subtle changes in stiffness and mechanical forces exerted on its environment (extracellular matrix (ECM)). Upon alteration of these biomechanical cues, endothelial cells initiate signaling pathways that govern vascular remodeling. The emerging organs-on-chip technologies allow the mimicking of complex microvasculature networks, identifying the combined or singular effects of these biomechanical or biochemical stimuli. Here, we present a microvasculature-on-chip model to investigate the singular effect of ECM stiffness and mechanical cyclic stretch on vascular development. Following two different approaches for vascular growth, the effect of ECM stiffness on sprouting angiogenesis and the effect of cyclic stretch on endothelial vasculogenesis are studied. Our results indicate that ECM hydrogel stiffness controls the size of the patterned vasculature and the density of sprouting angiogenesis. RNA sequencing shows that the cellular response to stretching is characterized by the upregulation of certain genes such as ANGPTL4+5, PDE1A, and PLEC.
Collapse
Affiliation(s)
- Dario Ferrari
- Organs-on-chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Arunima Sengupta
- Organs-on-chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Lyong Heo
- Stanford Center for Genomics and Personalized Medicine, Palo Alto, CA, USA
| | - Laszlo Pethö
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Laboratory for Mechanics of Materials and Nanostructures, Thun, Switzerland
| | - Johann Michler
- Empa, Swiss Federal Laboratories for Materials Science and Technology, Laboratory for Mechanics of Materials and Nanostructures, Thun, Switzerland
| | - Thomas Geiser
- Department of Pulmonary Medicine, Inselspital, University Hospital of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Vinicio A. de Jesus Perez
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University Medical Center, Stanford, CA, USA
| | - Wolfgang M. Kuebler
- Institute of Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Soheila Zeinali
- Organs-on-chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Olivier T. Guenat
- Organs-on-chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
- Department of Pulmonary Medicine, Inselspital, University Hospital of Bern, Bern, Switzerland
- Department of General Thoracic Surgery, Inselspital, University Hospital of Bern, Bern, Switzerland
| |
Collapse
|
22
|
Park HS, Lee JS, Kim C, Lee K, Hong I, Jung H, Lee H, Lee YJ, Ajiteru O, Sultan MT, Lee OJ, Kim SH, Park CH. Fluidic integrated 3D bioprinting system to sustain cell viability towards larynx fabrication. Bioeng Transl Med 2023; 8:e10423. [PMID: 36925698 PMCID: PMC10013754 DOI: 10.1002/btm2.10423] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 09/23/2022] [Accepted: 10/03/2022] [Indexed: 11/10/2022] Open
Abstract
Herein, we report the first study to create a three-dimensional (3D) bioprinted artificial larynx for whole-laryngeal replacement. Our 3D bio-printed larynx was generated using extrusion-based 3D bioprinter with rabbit's chondrocyte-laden gelatin methacryloyl (GelMA)/glycidyl-methacrylated hyaluronic acid (GMHA) hybrid bioink. We used a polycaprolactone (PCL) outer framework incorporated with pores to achieve the structural strength of printed constructs, as well as to provide a suitable microenvironment to support printed cells. Notably, we established a novel fluidics supply (FS) system that simultaneously supplies basal medium together with a 3D bioprinting process, thereby improving cell survival during the printing process. Our results showed that the FS system enhanced post-printing cell viability, which enabled the generation of a large-scale cell-laden artificial laryngeal framework. Additionally, the incorporation of the PCL outer framework with pores and inner hydrogel provides structural stability and sufficient nutrient/oxygen transport. An animal study confirmed that the transplanted 3D bio-larynx successfully maintained the airway. With further development, our new strategy holds great potential for fabricating human-scale larynxes with in vivo-like biological functions for laryngectomy patients.
Collapse
Affiliation(s)
- Hae Sang Park
- Department of Otorhinolaryngology–Head and Neck Surgery, Chuncheon Sacred Heart Hospital, College of MedicineHallym UniversityChuncheonRepublic of Korea
- Nano‐Bio Regenerative Medical Institute, School of MedicineHallym UniversityChuncheonRepublic of Korea
- Institute of New Frontier Research TeamHallym University, Hallym Clinical and Translation Science InstituteChuncheonRepublic of Korea
| | - Ji Seung Lee
- Nano‐Bio Regenerative Medical Institute, School of MedicineHallym UniversityChuncheonRepublic of Korea
| | - Chang‐Beom Kim
- Intelligent Robot Research TeamElectronics and Telecommunications Research InstituteDaejeonRepublic of Korea
| | - Kwang‐Ho Lee
- Department of Advanced Materials Science and Engineering, College of EngineeringKangwon National UniversityChuncheonRepublic of Korea
| | - In‐Sun Hong
- Department of Molecular Medicine, School of MedicineGachon UniversityIncheonRepublic of Korea
| | - Harry Jung
- Institute of New Frontier Research TeamHallym University, Hallym Clinical and Translation Science InstituteChuncheonRepublic of Korea
| | - Hanna Lee
- Nano‐Bio Regenerative Medical Institute, School of MedicineHallym UniversityChuncheonRepublic of Korea
| | - Young Jin Lee
- Nano‐Bio Regenerative Medical Institute, School of MedicineHallym UniversityChuncheonRepublic of Korea
| | - Olatunji Ajiteru
- Nano‐Bio Regenerative Medical Institute, School of MedicineHallym UniversityChuncheonRepublic of Korea
| | - Md Tipu Sultan
- Nano‐Bio Regenerative Medical Institute, School of MedicineHallym UniversityChuncheonRepublic of Korea
| | - Ok Joo Lee
- Nano‐Bio Regenerative Medical Institute, School of MedicineHallym UniversityChuncheonRepublic of Korea
| | - Soon Hee Kim
- Nano‐Bio Regenerative Medical Institute, School of MedicineHallym UniversityChuncheonRepublic of Korea
| | - Chan Hum Park
- Department of Otorhinolaryngology–Head and Neck Surgery, Chuncheon Sacred Heart Hospital, College of MedicineHallym UniversityChuncheonRepublic of Korea
- Nano‐Bio Regenerative Medical Institute, School of MedicineHallym UniversityChuncheonRepublic of Korea
| |
Collapse
|
23
|
Lee SR, Kim Y, Kim S, Kim J, Park S, Rhee S, Park D, Lee B, Baek K, Kim HY, Jeon NL. U-IMPACT: a universal 3D microfluidic cell culture platform. MICROSYSTEMS & NANOENGINEERING 2022; 8:126. [PMID: 36478874 PMCID: PMC9719897 DOI: 10.1038/s41378-022-00431-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/15/2022] [Accepted: 05/22/2022] [Indexed: 06/17/2023]
Abstract
The development of organs-on-a-chip has resulted in advances in the reconstruction of 3D cellular microenvironments. However, there remain limitations regarding applicability and manufacturability. Here, we present an injection-molded plastic array 3D universal culture platform (U-IMPACT) for various biological applications in a single platform, such as cocultures of various cell types, and spheroids (e.g., tumor spheroids, neurospheres) and tissues (e.g., microvessels). The U-IMPACT consists of three channels and a spheroid zone with a 96-well plate form factor. Specifically, organoids or spheroids (~500 μm) can be located in designated areas, while cell suspensions or cell-laden hydrogels can be selectively placed in three channels. For stable multichannel patterning, we developed a new patterning method based on capillary action, utilizing capillary channels and the native contact angle of the materials without any modification. We derived the optimal material hydrophilicity (contact angle of the body, 45-90°; substrate, <30°) for robust patterning through experiments and theoretical calculations. We demonstrated that the U-IMPACT can implement 3D tumor microenvironments for angiogenesis, vascularization, and tumor cell migration. Furthermore, we cultured neurospheres from induced neural stem cells. The U-IMPACT can serve as a multifunctional organ-on-a-chip platform for high-content and high-throughput screening.
Collapse
Affiliation(s)
- Seung-Ryeol Lee
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea
| | - Youngtaek Kim
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea
| | - Suryong Kim
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea
| | - Jiho Kim
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea
| | - Seonghyuk Park
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea
| | - Stephen Rhee
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea
| | - Dohyun Park
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea
| | | | | | - Ho-Young Kim
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea
- Institute of Advanced Machines and Design Seoul National University, Seoul, Republic of Korea
| | - Noo Li Jeon
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea
- Institute of Advanced Machines and Design Seoul National University, Seoul, Republic of Korea
- Institute of Bioengineering, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
24
|
Deng P, Zhao M, Zhang X, Qin J. A Transwell-Based Vascularized Model to Investigate the Effect of Interstitial Flow on Vasculogenesis. Bioengineering (Basel) 2022; 9:668. [PMID: 36354579 PMCID: PMC9687519 DOI: 10.3390/bioengineering9110668] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/27/2022] [Accepted: 10/30/2022] [Indexed: 09/08/2024] Open
Abstract
Interstitial flow plays a significant role in vascular system development, mainly including angiogenesis and vasculogenesis. However, compared to angiogenesis, the effect of interstitial flow on vasculogenesis is less explored. Current in vitro models for investigating the effect of interstitial flow on vasculogenesis heavily rely on microfluidic chips, which require microfluidic expertise and facilities, and may not be accessible to biological labs. Here, we proposed a facile approach to building perfusable vascular networks through the self-assembly of endothelial cells in a modified transwell format and investigated the effect of interstitial flow on vasculogenesis. We found that the effect of interstitial flow on vasculogenesis was closely related to the existence of VEGF and fibroblasts in the developed model: (1) In the presence of fibroblasts, interstitial flow (within the range of 0.1-0.6 μm/s) facilitated the perfusability of the engineered vasculatures. Additional VEGF in the culture medium further worked synergically with interstitial flow to develop longer, wider, denser, and more perfusable vasculatures than static counterparts; (2) In the absence of fibroblasts, vasculatures underwent severe regression within 7 days under static conditions. However, interstitial flow greatly inhibited vessel regression and enhanced vascular perfusability and morphogenesis without the need for additional VEGF. These results revealed that the effect of interstitial flow might vary depending on the existence of VEGF and fibroblasts, and would provide some guidelines for constructing in vitro self-assembled vasculatures. The established transwell-based vascularized model provides a simple method to build perfusable vasculatures and could also be utilized for creating functional tissues in regenerative medicine.
Collapse
Affiliation(s)
- Pengwei Deng
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mengqian Zhao
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xu Zhang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Jianhua Qin
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
25
|
Sethakorn N, Heninger E, Breneman MT, Recchia E, Ding AB, Jarrard DF, Hematti P, Beebe DJ, Kosoff D. Integrated analysis of the tumor microenvironment using a reconfigurable microfluidic cell culture platform. FASEB J 2022; 36:e22540. [PMID: 36083096 PMCID: PMC9476232 DOI: 10.1096/fj.202200684rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/22/2022] [Accepted: 08/29/2022] [Indexed: 11/11/2022]
Abstract
The tumor microenvironment (TME) is a complex network of non-malignant cells and stroma that perform a wide array of vital roles in tumor growth, immune evasion, metastasis, and therapeutic resistance. These highly diverse roles have been shown to be critically important to the progression of cancers and have already shown potential as therapeutic targets. Therefore, there has been a tremendous push to elucidate the pathways that underlie these roles and to develop new TME-directed therapies for cancer treatment. Unfortunately, TME-focused research has been limited by a lack of translational in vitro culture platforms that can model this highly complex niche and can support the integrated analysis of cell biology and function. In the current study, we investigate whether an independently developed reconfigurable microfluidic platform, known as Stacks, can address the critical need for translational multi-cellular tumor models and integrated analytics in TME research. We present data on multi-cellular culture of primary human cells in Stacks as well as the orthogonal analysis of cellular polarization, differentiation, migration, and cytotoxicity in this reconfigurable system. These expanded capabilities of Stacks are highly relevant to the cancer research community with the potential to enhance clinical translation of pre-clinical TME studies and to yield novel biological insight into TME crosstalk, metastasis, and responses to novel drug combinations or immune therapies.
Collapse
Affiliation(s)
- Nan Sethakorn
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Erika Heninger
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Matthew T Breneman
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Emma Recchia
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Adeline B Ding
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - David F Jarrard
- Department of Urology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Peiman Hematti
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - David J Beebe
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - David Kosoff
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA.,William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| |
Collapse
|
26
|
Simitian G, Virumbrales-Muñoz M, Sánchez-de-Diego C, Beebe DJ, Kosoff D. Microfluidics in vascular biology research: a critical review for engineers, biologists, and clinicians. LAB ON A CHIP 2022; 22:3618-3636. [PMID: 36047330 PMCID: PMC9530010 DOI: 10.1039/d2lc00352j] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Neovascularization, the formation of new blood vessels, has received much research attention due to its implications for physiological processes and diseases. Most studies using traditional in vitro and in vivo platforms find challenges in recapitulating key cellular and mechanical cues of the neovascularization processes. Microfluidic in vitro models have been presented as an alternative to these limitations due to their capacity to leverage microscale physics to control cell organization and integrate biochemical and mechanical cues, such as shear stress, cell-cell interactions, or nutrient gradients, making them an ideal option for recapitulating organ physiology. Much has been written about the use of microfluidics in vascular biology models from an engineering perspective. However, a review introducing the different models, components and progress for new potential adopters of these technologies was absent in the literature. Therefore, this paper aims to approach the use of microfluidic technologies in vascular biology from a perspective of biological hallmarks to be studied and written for a wide audience ranging from clinicians to engineers. Here we review applications of microfluidics in vascular biology research, starting with design considerations and fabrication techniques. After that, we review the state of the art in recapitulating angiogenesis and vasculogenesis, according to the hallmarks recapitulated and complexity of the models. Finally, we discuss emerging research areas in neovascularization, such as drug discovery, and potential future directions.
Collapse
Affiliation(s)
- Grigor Simitian
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA.
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - María Virumbrales-Muñoz
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Cristina Sánchez-de-Diego
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - David J Beebe
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - David Kosoff
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA.
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
27
|
Terrassoux L, Claux H, Bacari S, Meignan S, Furlan A. A Bloody Conspiracy. Blood Vessels and Immune Cells in the Tumor Microenvironment. Cancers (Basel) 2022; 14:cancers14194581. [PMID: 36230504 PMCID: PMC9558972 DOI: 10.3390/cancers14194581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/10/2022] [Accepted: 09/15/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary The tumor microenvironment has risen over the last years as a significant contributor to the failure of antitumoral strategies due to its numerous pro-tumorigenic activities. In this review, we focused on two features of this microenvironment, namely angiogenesis and immunity, which have been the targets of therapies to tackle tumors via its microenvironmental part over the last decade. Increasing our knowledge of the complex interactions within this ecosystem is mandatory to optimize these therapeutic approaches. The development of innovative experimental models is of great help in reaching this goal. Abstract Cancer progression occurs in concomitance with a profound remodeling of the cellular microenvironment. Far from being a mere passive event, the re-orchestration of interactions between the various cell types surrounding tumors highly contributes to the progression of the latter. Tumors notably recruit and stimulate the sprouting of new blood vessels through a process called neo-angiogenesis. Beyond helping the tumor cope with an increased metabolic demand associated with rapid growth, this also controls the metastatic dissemination of cancer cells and the infiltration of immune cells in the tumor microenvironment. To decipher this critical interplay for the clinical progression of tumors, the research community has developed several valuable models in the last decades. This review offers an overview of the various instrumental solutions currently available, including microfluidic chips, co-culture models, and the recent rise of organoids. We highlight the advantages of each technique and the specific questions they can address to better understand the tumor immuno-angiogenic ecosystem. Finally, we discuss this development field’s fundamental and applied perspectives.
Collapse
Affiliation(s)
- Lisa Terrassoux
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
| | - Hugo Claux
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
| | - Salimata Bacari
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
| | - Samuel Meignan
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
| | - Alessandro Furlan
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
- Correspondence:
| |
Collapse
|
28
|
Lai X, Yang M, Wu H, Li D. Modular Microfluidics: Current Status and Future Prospects. MICROMACHINES 2022; 13:1363. [PMID: 36014285 PMCID: PMC9414757 DOI: 10.3390/mi13081363] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 06/15/2023]
Abstract
This review mainly studies the development status, limitations, and future directions of modular microfluidic systems. Microfluidic technology is an important tool platform for scientific research and plays an important role in various fields. With the continuous development of microfluidic applications, conventional monolithic microfluidic chips show more and more limitations. A modular microfluidic system is a system composed of interconnected, independent modular microfluidic chips, which are easy to use, highly customizable, and on-site deployable. In this paper, the current forms of modular microfluidic systems are classified and studied. The popular fabrication techniques for modular blocks, the major application scenarios of modular microfluidics, and the limitations of modular techniques are also discussed. Lastly, this review provides prospects for the future direction of modular microfluidic technologies.
Collapse
Affiliation(s)
- Xiaochen Lai
- School of Automation, Nanjing University of Information Science & Technology, Nanjing 210044, China
| | - Mingpeng Yang
- School of Automation, Nanjing University of Information Science & Technology, Nanjing 210044, China
| | - Hao Wu
- State Key Laboratory of Precision Measuring Technology and Instruments, Tianjin University, Tianjin 300072, China
| | - Dachao Li
- State Key Laboratory of Precision Measuring Technology and Instruments, Tianjin University, Tianjin 300072, China
| |
Collapse
|
29
|
Marzagalli M, Pelizzoni G, Fedi A, Vitale C, Fontana F, Bruno S, Poggi A, Dondero A, Aiello M, Castriconi R, Bottino C, Scaglione S. A multi-organ-on-chip to recapitulate the infiltration and the cytotoxic activity of circulating NK cells in 3D matrix-based tumor model. Front Bioeng Biotechnol 2022; 10:945149. [PMID: 35957642 PMCID: PMC9358021 DOI: 10.3389/fbioe.2022.945149] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 06/29/2022] [Indexed: 11/22/2022] Open
Abstract
The success of immunotherapeutic approaches strictly depends on the immune cells interaction with cancer cells. While conventional in vitro cell cultures under-represent the complexity and dynamic crosstalk of the tumor microenvironment, animal models do not allow deciphering the anti-tumor activity of the human immune system. Therefore, the development of reliable and predictive preclinical models has become crucial for the screening of immune-therapeutic approaches. We here present an organ-on-chip organ on chips (OOC)-based approach for recapitulating the immune cell Natural Killer (NK) migration under physiological fluid flow, infiltration within a 3D tumor matrix, and activation against neuroblastoma cancer cells in a humanized, fluid-dynamic environment. Circulating NK cells actively initiate a spontaneous “extravasation” process toward the physically separated tumor niche, retaining their ability to interact with matrix-embedded tumor cells, and to display a cytotoxic effect (tumor cell apoptosis). Since NK cells infiltration and phenotype is correlated with prognosis and response to immunotherapy, their phenotype is also investigated: most importantly, a clear decrease in CD16-positive NK cells within the migrated and infiltrated population is observed. The proposed immune-tumor OOC-based model represents a promising approach for faithfully recapitulating the human pathology and efficiently employing the immunotherapies testing, eventually in a personalized perspective. An immune-organ on chip to recapitulate the tumor-mediated infiltration of circulating immune cells within 3D tumor model.
Collapse
Affiliation(s)
| | - Giorgia Pelizzoni
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, Piazza Della Scienza, Milan, Italy
| | - Arianna Fedi
- National Research Council, CNR-IEIIT, Genoa, Italy
| | - Chiara Vitale
- National Research Council, CNR-IEIIT, Genoa, Italy
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Fabrizio Fontana
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, Milan, Italy
| | - Silvia Bruno
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Alessandro Poggi
- Molecular Oncology and Angiogenesis Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Alessandra Dondero
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
- IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | - Roberta Castriconi
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
- IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Cristina Bottino
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
- IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Silvia Scaglione
- National Research Council, CNR-IEIIT, Genoa, Italy
- *Correspondence: Silvia Scaglione,
| |
Collapse
|
30
|
Karkampouna S, Kruithof-de Julio M, Thalmann GN. Role of prostate and bone stromal cells on prostate cancer progression. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2022; 10:180-187. [PMID: 35874291 PMCID: PMC9301059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 04/14/2022] [Indexed: 06/15/2023]
Abstract
Prostate cancer is a highly heterogeneous disease, often manifesting in a metastatic form to the bone. Complex tumour cells and surrounding microenvironment interactions are important determinants of disease progression and therapy resistance. Here, we provide an overview of some of the early studies that recognized the pro-tumourigenic role of prostate stroma, particularly fibroblasts, bone stromal components, and its permissive tumour properties. This article is written in memory of Prof. Dr LWK Chung and his contributions. Prostate and bone metastasis stroma concepts emerging from his work are now more actively being pursued by the authors of this article and others in the field.
Collapse
Affiliation(s)
- Sofia Karkampouna
- Department for BioMedical Research DBMR, Urology Research Laboratory, University of BernBern, Switzerland
| | - Marianna Kruithof-de Julio
- Department for BioMedical Research DBMR, Urology Research Laboratory, University of BernBern, Switzerland
- Translational Organoid Resource, Department for BioMedical Research, University of BernBern, Switzerland
- Bern Center for Precision Medicine, University of Bern and InselspitalBern, Switzerland
- Department of Urology, Inselspital, Bern University HospitalBern, Switzerland
| | - George N Thalmann
- Department for BioMedical Research DBMR, Urology Research Laboratory, University of BernBern, Switzerland
- Department of Urology, Inselspital, Bern University HospitalBern, Switzerland
| |
Collapse
|
31
|
Filippi M, Buchner T, Yasa O, Weirich S, Katzschmann RK. Microfluidic Tissue Engineering and Bio-Actuation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2108427. [PMID: 35194852 DOI: 10.1002/adma.202108427] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/07/2022] [Indexed: 06/14/2023]
Abstract
Bio-hybrid technologies aim to replicate the unique capabilities of biological systems that could surpass advanced artificial technologies. Soft bio-hybrid robots consist of synthetic and living materials and have the potential to self-assemble, regenerate, work autonomously, and interact safely with other species and the environment. Cells require a sufficient exchange of nutrients and gases, which is guaranteed by convection and diffusive transport through liquid media. The functional development and long-term survival of biological tissues in vitro can be improved by dynamic flow culture, but only microfluidic flow control can develop tissue with fine structuring and regulation at the microscale. Full control of tissue growth at the microscale will eventually lead to functional macroscale constructs, which are needed as the biological component of soft bio-hybrid technologies. This review summarizes recent progress in microfluidic techniques to engineer biological tissues, focusing on the use of muscle cells for robotic bio-actuation. Moreover, the instances in which bio-actuation technologies greatly benefit from fusion with microfluidics are highlighted, which include: the microfabrication of matrices, biomimicry of cell microenvironments, tissue maturation, perfusion, and vascularization.
Collapse
Affiliation(s)
- Miriam Filippi
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Thomas Buchner
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Oncay Yasa
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Stefan Weirich
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Robert K Katzschmann
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| |
Collapse
|
32
|
Ronaldson-Bouchard K, Baldassarri I, Tavakol DN, Graney PL, Samaritano M, Cimetta E, Vunjak-Novakovic G. Engineering complexity in human tissue models of cancer. Adv Drug Deliv Rev 2022; 184:114181. [PMID: 35278521 PMCID: PMC9035134 DOI: 10.1016/j.addr.2022.114181] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/15/2022] [Accepted: 03/04/2022] [Indexed: 02/06/2023]
Abstract
Major progress in the understanding and treatment of cancer have tremendously improved our knowledge of this complex disease and improved the length and quality of patients' lives. Still, major challenges remain, in particular with respect to cancer metastasis which still escapes effective treatment and remains responsible for 90% of cancer related deaths. In recent years, the advances in cancer cell biology, oncology and tissue engineering converged into the engineered human tissue models of cancer that are increasingly recapitulating many aspects of cancer progression and response to drugs, in a patient-specific context. The complexity and biological fidelity of these models, as well as the specific questions they aim to investigate, vary in a very broad range. When selecting and designing these experimental models, the fundamental question is "how simple is complex enough" to accomplish a specific goal of cancer research. Here we review the state of the art in developing and using the human tissue models in cancer research and developmental drug screening. We describe the main classes of models providing different levels of biological fidelity and complexity, discuss their advantages and limitations, and propose a framework for designing an appropriate model for a given study. We close by outlining some of the current needs, opportunities and challenges in this rapidly evolving field.
Collapse
Affiliation(s)
- Kacey Ronaldson-Bouchard
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA
| | - Ilaria Baldassarri
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA
| | - Daniel Naveed Tavakol
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA
| | - Pamela L Graney
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA
| | - Maria Samaritano
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA
| | - Elisa Cimetta
- Department of Industrial Engineering, University of Padua, Via Marzolo 9, 35131 Padova, Italy; Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Corso Stati Uniti 4, 35127 Padova, Italy
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA; Department of Medicine, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA; College of Dental Medicine, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA.
| |
Collapse
|
33
|
Silberman J, Jha A, Ryan H, Abbate T, Moore E. Modeled vascular microenvironments: immune-endothelial cell interactions in vitro. Drug Deliv Transl Res 2021; 11:2482-2495. [PMID: 33797034 DOI: 10.1007/s13346-021-00970-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2021] [Indexed: 10/21/2022]
Abstract
The advancement of in vitro techniques enables a better understanding of biological processes and improves drug screening platforms. In vitro studies allow for enhanced observation of cell behavior, control over the mimicked microenvironment, and the ability to use human cells. In particular, advances in vascular microenvironment recapitulation are of interest given vasculature influence in cardiovascular vascular diseases and cancer. These investigate alterations in endothelial cell behavior and immune cell interactions with endothelial cells. Specific immune cells such as monocytes, macrophages, neutrophils, and T cells influence endothelial cell behavior by promoting or inhibiting vasculogenesis through cell-cell interaction or soluble signaling. Results from these studies showcase cell behavior in vascular diseases and in the context of tumor metastasis. In this review, we discuss examples of in vitro studies modeling immune cell-endothelial cell interactions to present methods and recent findings in the field. Schematic showcasing common methods of in vitro experimentation of endothelial-immune cell interactions, including interactions with flow, static culture, or in-direct contact.
Collapse
Affiliation(s)
- Justin Silberman
- Materials Science and Engineering, University of Florida, FL, Gainesville, USA
| | - Aakanksha Jha
- Biomedical Engineering, University of Florida, FL, Gainesville, USA
| | - Holly Ryan
- Biomedical Engineering, University of Florida, FL, Gainesville, USA
| | - Talia Abbate
- Materials Science and Engineering, University of Florida, FL, Gainesville, USA
| | - Erika Moore
- Materials Science and Engineering, University of Florida, FL, Gainesville, USA.
- Biomedical Engineering, University of Florida, FL, Gainesville, USA.
| |
Collapse
|
34
|
Molloy A, Harrison J, McGrath JS, Owen Z, Smith C, Liu X, Li X, Cox JAG. Microfluidics as a Novel Technique for Tuberculosis: From Diagnostics to Drug Discovery. Microorganisms 2021; 9:microorganisms9112330. [PMID: 34835455 PMCID: PMC8618277 DOI: 10.3390/microorganisms9112330] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 12/29/2022] Open
Abstract
Tuberculosis (TB) remains a global healthcare crisis, with an estimated 5.8 million new cases and 1.5 million deaths in 2020. TB is caused by infection with the major human pathogen Mycobacterium tuberculosis, which is difficult to rapidly diagnose and treat. There is an urgent need for new methods of diagnosis, sufficient in vitro models that capably mimic all physiological conditions of the infection, and high-throughput drug screening platforms. Microfluidic-based techniques provide single-cell analysis which reduces experimental time and the cost of reagents, and have been extremely useful for gaining insight into monitoring microorganisms. This review outlines the field of microfluidics and discusses the use of this novel technique so far in M. tuberculosis diagnostics, research methods, and drug discovery platforms. The practices of microfluidics have promising future applications for diagnosing and treating TB.
Collapse
Affiliation(s)
- Antonia Molloy
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK; (A.M.); (J.H.)
| | - James Harrison
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK; (A.M.); (J.H.)
| | - John S. McGrath
- Sphere Fluidics Limited, The McClintock Building, Suite 7, Granta Park, Great Abington, Cambridge CB21 6GP, UK; (J.S.M.); (Z.O.); (C.S.); (X.L.); (X.L.)
| | - Zachary Owen
- Sphere Fluidics Limited, The McClintock Building, Suite 7, Granta Park, Great Abington, Cambridge CB21 6GP, UK; (J.S.M.); (Z.O.); (C.S.); (X.L.); (X.L.)
| | - Clive Smith
- Sphere Fluidics Limited, The McClintock Building, Suite 7, Granta Park, Great Abington, Cambridge CB21 6GP, UK; (J.S.M.); (Z.O.); (C.S.); (X.L.); (X.L.)
| | - Xin Liu
- Sphere Fluidics Limited, The McClintock Building, Suite 7, Granta Park, Great Abington, Cambridge CB21 6GP, UK; (J.S.M.); (Z.O.); (C.S.); (X.L.); (X.L.)
| | - Xin Li
- Sphere Fluidics Limited, The McClintock Building, Suite 7, Granta Park, Great Abington, Cambridge CB21 6GP, UK; (J.S.M.); (Z.O.); (C.S.); (X.L.); (X.L.)
| | - Jonathan A. G. Cox
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK; (A.M.); (J.H.)
- Correspondence: ; Tel.: +44-121-204-5011
| |
Collapse
|
35
|
Mohan MD, Young EWK. TANDEM: biomicrofluidic systems with transverse and normal diffusional environments for multidirectional signaling. LAB ON A CHIP 2021; 21:4081-4094. [PMID: 34604885 DOI: 10.1039/d1lc00279a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Biomicrofluidic systems that can recapitulate complex biological processes with precisely controlled 3D geometries are a significant advancement from traditional 2D cultures. To this point, these systems have largely been limited to either laterally adjacent channels in a single plane or vertically stacked single-channel arrangements. As a result, lateral (or transverse) and vertical (or normal) diffusion have been isolated to their respective designs only, thus limiting potential access to nutrients and 3D communication that typifies in vivo microenvironments. Here we report a novel device architecture called "TANDEM", an acronym for "T̲ransverse A̲nd N̲ormal D̲iffusional E̲nvironments for M̲ultidirectional Signaling", which enables multiplanar arrangements of aligned channels where normal and transverse diffusion occur in tandem to facilitate multidirectional communication. We developed a computational transport model in COMSOL and tested diffusion and culture viability in one specific TANDEM configuration, and found that TANDEM systems demonstrated enhanced diffusion in comparison to single-plane counterparts. This resulted in improved viability of hydrogel-embedded cells, which typically suffer from a lack of sufficient nutrient access during long-term culture. Finally, we showed that TANDEM designs can be expanded to more complex alternative configurations depending on the needs of the end-user. Based on these findings, TANDEM designs can utilize multidirectional enhanced diffusion to improve long-term viability and ultimately facilitate more robust and more biomimetic microfluidic systems with increasingly more complex geometric layouts.
Collapse
Affiliation(s)
- Michael D Mohan
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON, M5S 3G8, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada
| | - Edmond W K Young
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON, M5S 3G8, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada
| |
Collapse
|
36
|
Heninger E, Kosoff D, Rodems TS, Sethakorn N, Singh A, Gungurthi H, Carlson KN, Yang B, Gilsdorf C, Pasch CA, Deming DA, Ellis L, Beebe DJ, Jarrard DF, Lang JM. Live cell molecular analysis of primary prostate cancer organoids identifies persistent androgen receptor signaling. Med Oncol 2021; 38:135. [PMID: 34581895 PMCID: PMC8478748 DOI: 10.1007/s12032-021-01582-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 09/12/2021] [Indexed: 11/29/2022]
Abstract
Prostate Cancer (PC) is a disease with remarkable tumor heterogeneity that often manifests in significant intra-patient variability with regards to clinical outcomes and treatment response. Commonly available PC cell lines do not accurately reflect the complexity of this disease and there is critical need for development of new models to recapitulate the intricate hierarchy of tumor pathogenesis. In current study, we established ex vivo primary patient-derived cancer organoid (PDCO) cultures from prostatectomy specimens of patients with locally advanced PC. We then performed a comprehensive multi-parameter characterization of the cellular composition utilizing a novel approach for live-cell staining and direct imaging in the integrated microfluidic Stacks device. Using orthogonal flow cytometry analysis, we demonstrate that primary PDCOs maintain distinct subsets of epithelial cells throughout culture and that these cells conserve expression of androgen receptor (AR)-related elements. Furthermore, to confirm the tumor-origin of the PDCOs we have analyzed the expression of PC-associated epigenetic biomarkers including promoter methylation of the GSTP1, RASSF1 and APC and RARb genes by employing a novel microfluidic rare-event screening protocol. These results demonstrate that this ex vivo PDCO model recapitulates the complexity of the epithelial tumor microenvironment of multifocal PC using orthogonal analyses. Furthermore, we propose to leverage the Stacks microfluidic device as a high-throughput, translational platform to interrogate phenotypic and molecular endpoints with the capacity to incorporate a complex tumor microenvironment.
Collapse
Affiliation(s)
- Erika Heninger
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave., Madison, USA
| | - David Kosoff
- Department of Medicine, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Tamara S Rodems
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave., Madison, USA
| | - Nan Sethakorn
- Department of Medicine, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Anupama Singh
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave., Madison, USA
| | - Harshitha Gungurthi
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave., Madison, USA
| | - Kristin N Carlson
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave., Madison, USA
| | - Bing Yang
- Department of Urology, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Cole Gilsdorf
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave., Madison, USA
| | - Cheri A Pasch
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave., Madison, USA
| | - Dustin A Deming
- Department of Medicine, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Leigh Ellis
- Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, Samuel Oschin Comprehensive Cancer Institute, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - David J Beebe
- Department of Pathology and Laboratory Medicine, 1111 Highland Ave., Madison, WI, 53705, USA
| | - David F Jarrard
- Department of Urology, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Joshua M Lang
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave., Madison, USA. .,Department of Medicine, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA. .,Wisconsin Institutes for Medical Research, Rm 7151, 1111 Highland Ave., Madison, WI, 53705, USA.
| |
Collapse
|
37
|
Ando Y, Mariano C, Shen K. Engineered in vitro tumor models for cell-based immunotherapy. Acta Biomater 2021; 132:345-359. [PMID: 33857692 PMCID: PMC8434941 DOI: 10.1016/j.actbio.2021.03.076] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 03/26/2021] [Accepted: 03/30/2021] [Indexed: 12/15/2022]
Abstract
Tumor immunotherapy is rapidly evolving as one of the major pillars of cancer treatment. Cell-based immunotherapies, which utilize patient's own immune cells to eliminate cancer cells, have shown great promise in treating a range of malignancies, especially those of hematopoietic origins. However, their performance on a broader spectrum of solid tumor types still fall short of expectations in the clinical stage despite promising preclinical assessments. In this review, we briefly introduce cell-based immunotherapies and the inhibitory mechanisms in tumor microenvironments that may have contributed to this discrepancy. Specifically, a major obstacle to the clinical translation of cell-based immunotherapies is in the lack of preclinical models that can accurately assess the efficacies and mechanisms of these therapies in a (patho-)physiologically relevant manner. Lately, tissue engineering and organ-on-a-chip tools and microphysiological models have allowed for more faithful recapitulation of the tumor microenvironments, by incorporating crucial tumor tissue features such as cellular phenotypes, tissue architecture, extracellular matrix, physical parameters, and their dynamic interactions. This review summarizes the existing engineered tumor models with a focus on tumor immunology and cell-based immunotherapy. We also discuss some key considerations for the future development of engineered tumor models for immunotherapeutics. STATEMENT OF SIGNIFICANCE: Cell-based immunotherapies have shown great promise in treating hematological malignancies and some epithelial tumors. However, their performance on a broader spectrum of solid tumor types still fall short of expectations. Major obstacles include the inhibitory mechanisms in tumor microenvironments (TME) and the lack of preclinical models that can accurately assess the efficacies and mechanisms of cellular therapies in a (patho-)physiologically relevant manner. In this review, we introduce recent progress in tissue engineering and microphysiological models for more faithful recapitulation of TME for cell-based immunotherapies, and some key considerations for the future development of engineered tumor models. This overview will provide a better understanding on the role of engineered models in accelerating immunotherapeutic discoveries and clinical translations.
Collapse
Affiliation(s)
- Yuta Ando
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, United States
| | - Chelsea Mariano
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, United States
| | - Keyue Shen
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, United States; Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, United States; USC Stem Cell, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States.
| |
Collapse
|
38
|
Del Piccolo N, Shirure VS, Bi Y, Goedegebuure SP, Gholami S, Hughes CC, Fields RC, George SC. Tumor-on-chip modeling of organ-specific cancer and metastasis. Adv Drug Deliv Rev 2021; 175:113798. [PMID: 34015419 DOI: 10.1016/j.addr.2021.05.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/04/2021] [Accepted: 05/11/2021] [Indexed: 02/08/2023]
Abstract
Every year, cancer claims millions of lives around the globe. Unfortunately, model systems that accurately mimic human oncology - a requirement for the development of more effective therapies for these patients - remain elusive. Tumor development is an organ-specific process that involves modification of existing tissue features, recruitment of other cell types, and eventual metastasis to distant organs. Recently, tissue engineered microfluidic devices have emerged as a powerful in vitro tool to model human physiology and pathology with organ-specificity. These organ-on-chip platforms consist of cells cultured in 3D hydrogels and offer precise control over geometry, biological components, and physiochemical properties. Here, we review progress towards organ-specific microfluidic models of the primary and metastatic tumor microenvironments. Despite the field's infancy, these tumor-on-chip models have enabled discoveries about cancer immunobiology and response to therapy. Future work should focus on the development of autologous or multi-organ systems and inclusion of the immune system.
Collapse
|
39
|
Zhang Q, Feng S, Lin L, Mao S, Lin JM. Emerging open microfluidics for cell manipulation. Chem Soc Rev 2021; 50:5333-5348. [PMID: 33972984 DOI: 10.1039/d0cs01516d] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cell manipulation is the foundation of biochemical studies, which demands user-friendly, multifunctional and precise tools. Based on flow confinement principles, open microfluidics can control the movement of microscale liquid in open space. Every position of the circuit is accessible to external instruments, making it possible to perform precise treatment and analysis of cells at arbitrary target positions especially at the single-cell/sub-cell level. Benefiting from its unique superiority, various manipulations including patterned cell culture, 3D tissue modelling, localized chemical stimulation, online cellular factor analysis, single cell sampling, partial cell treatment, and subcellular free radical attack can be easily realized. In this tutorial review, we summarize two basic ideas to design open microfluidics: open microfluidic networks and probes. The principles of mainstream open microfluidic methods are explained, and their recent important applications are introduced. Challenges and developing trends of open microfluidics are also discussed.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, 100084, China.
| | - Shuo Feng
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, 100084, China.
| | - Ling Lin
- Department of Bioengineering, Beijing Technology and Business University, Beijing 100048, China.
| | - Sifeng Mao
- Department of Applied Chemistry, Graduate School of Urban Environmental Sciences, Tokyo Metropolitan University, Minamiohsawa, Hachioji, Tokyo 192-0397, Japan
| | - Jin-Ming Lin
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
40
|
Maulana TI, Kromidas E, Wallstabe L, Cipriano M, Alb M, Zaupa C, Hudecek M, Fogal B, Loskill P. Immunocompetent cancer-on-chip models to assess immuno-oncology therapy. Adv Drug Deliv Rev 2021; 173:281-305. [PMID: 33798643 DOI: 10.1016/j.addr.2021.03.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 03/08/2021] [Accepted: 03/17/2021] [Indexed: 12/12/2022]
Abstract
The advances in cancer immunotherapy come with several obstacles, limiting its widespread use and benefits so far only to a small subset of patients. One of the underlying challenges remains to be the lack of representative nonclinical models that translate to human immunity and are able to predict clinical efficacy and safety outcomes. In recent years, immunocompetent Cancer-on-Chip models emerge as an alternative human-based platform that enables the integration and manipulation of complex tumor microenvironment. In this review, we discuss novel opportunities offered by Cancer-on-Chip models to advance (mechanistic) immuno-oncology research, ranging from design flexibility to multimodal analysis approaches. We then exemplify their (potential) applications for the research and development of adoptive cell therapy, immune checkpoint therapy, cytokine therapy, oncolytic virus, and cancer vaccines.
Collapse
|
41
|
Richardson IM, Calo CJ, Hind LE. Microphysiological Systems for Studying Cellular Crosstalk During the Neutrophil Response to Infection. Front Immunol 2021; 12:661537. [PMID: 33986752 PMCID: PMC8111168 DOI: 10.3389/fimmu.2021.661537] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 04/09/2021] [Indexed: 12/13/2022] Open
Abstract
Neutrophils are the primary responders to infection, rapidly migrating to sites of inflammation and clearing pathogens through a variety of antimicrobial functions. This response is controlled by a complex network of signals produced by vascular cells, tissue resident cells, other immune cells, and the pathogen itself. Despite significant efforts to understand how these signals are integrated into the neutrophil response, we still do not have a complete picture of the mechanisms regulating this process. This is in part due to the inherent disadvantages of the most-used experimental systems: in vitro systems lack the complexity of the tissue microenvironment and animal models do not accurately capture the human immune response. Advanced microfluidic devices incorporating relevant tissue architectures, cell-cell interactions, and live pathogen sources have been developed to overcome these challenges. In this review, we will discuss the in vitro models currently being used to study the neutrophil response to infection, specifically in the context of cell-cell interactions, and provide an overview of their findings. We will also provide recommendations for the future direction of the field and what important aspects of the infectious microenvironment are missing from the current models.
Collapse
Affiliation(s)
| | | | - Laurel E. Hind
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO, United States
| |
Collapse
|
42
|
Kim S, Ko J, Lee SR, Park D, Park S, Jeon NL. Anchor-IMPACT: A standardized microfluidic platform for high-throughput antiangiogenic drug screening. Biotechnol Bioeng 2021; 118:2524-2535. [PMID: 33764506 DOI: 10.1002/bit.27765] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/16/2021] [Accepted: 03/23/2021] [Indexed: 01/22/2023]
Abstract
In vitro models are becoming more advanced to truly present physiological systems while enabling high-throughput screening and analysis. Organ-on-a-chip devices provide remarkable results through the reconstruction of a three-dimensional (3D) cellular microenvironment although they need to be further developed in terms of multiple liquid patterning principle, material properties, and scalability. Here we present a 3D anchor-based microfluidic injection-molded plastic array culture platform (Anchor-IMPACT) that enables selective, space-intensive patterning of hydrogels using anchor-island for high-throughput angiogenesis evaluation model. Anchor-IMPACT consists of a central channel and an anchor-island, integrating the array into an abbreviated 96-well plate format with a standard microscope slide size. The anchor-island enables selective 3D cell patterning without channel-to-channel contact or any hydrogel injection port using an anchor structure unlike conventional culture compartment. The hydrogel was patterned into defined regions by spontaneous capillary flow under hydrophilic conditions. We configured multiple cell patterning structures to investigate the angiogenic potency of colorectal cancer cells in Anchor-IMPACT and the morphological properties of the angiogenesis induced by the paracrine effect were evaluated. In addition, the efficacy of anticancer drugs against angiogenic sprouts was verified by following dose-dependent responses. Our results indicate that Anchor-IMPACT offers not only a model of high-throughput experimentation but also an advanced 3D cell culture platform and can significantly improve current in vitro models while providing the basis for developing predictive preclinical models for biopharmaceutical applications.
Collapse
Affiliation(s)
- Suryong Kim
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea
| | - Jihoon Ko
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea
| | - Seung-Ryeol Lee
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea
| | - Dohyun Park
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea
| | - Seunghyuk Park
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea
| | - Noo Li Jeon
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea.,Institute of Advanced Machines and Design, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
43
|
Ahmed A, Joshi IM, Larson S, Mansouri M, Gholizadeh S, Allahyari Z, Forouzandeh F, Borkholder DA, Gaborski TR, Abhyankar VV. Microengineered 3D Collagen Gels with Independently Tunable Fiber Anisotropy and Directionality. ADVANCED MATERIALS TECHNOLOGIES 2021; 6:2001186. [PMID: 34150990 PMCID: PMC8211114 DOI: 10.1002/admt.202001186] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Indexed: 05/17/2023]
Abstract
Cellular processes, including differentiation, proliferation, and migration, have been linked to the alignment (anisotropy) and orientation (directionality) of collagen fibers in the native extracellular matrix (ECM). Given the critical role that biophysical cell-matrix interactions play in regulating biological functions, several microfluidic-based methods have been used to establish 3D collagen gels with defined fiber properties; these gels have helped to establish quantitative relationships between structural ECM cues and observed cell responses. Although existing microfluidic fabrication methods provide excellent definition over collagen fiber anisotropy, they have not demonstrated the independent control over fiber anisotropy and directionality necessary to replicate in vivo collagen architecture. Therefore, to advance collagen microengineering capabilities, we present a user-friendly technology platform that uses controlled fluid flows within a non-uniform microfluidic channel network to create collagen landscapes that can be tuned as a function of extensional strain rate. Herein, we demonstrate capabilities to i) control the degree of fiber anisotropy, ii) create spatial gradients in fiber anisotropy, iii) independently define fiber directionality, and iv) generate multi-material interfaces within a 3D environment. We then address the practical issue of integrating cells into microfluidic systems by using a peel-off template technique to provide direct access to microengineered collagen gels, and demonstrate that cells respond to the defined properties of the landscape. Finally, the platform's modular capability is highlighted by integrating a sub-micrometer thick porous parylene membrane onto the microengineered collagen as a method to define cell-substrate interactions.
Collapse
Affiliation(s)
- Adeel Ahmed
- Microsystems Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Indranil M Joshi
- Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Stephen Larson
- Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Mehran Mansouri
- Microsystems Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Shayan Gholizadeh
- Microsystems Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Zahra Allahyari
- Microsystems Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Farzad Forouzandeh
- Microsystems Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - David A Borkholder
- Microsystems Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Thomas R Gaborski
- Microsystems Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
- Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Vinay V Abhyankar
- Microsystems Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
- Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| |
Collapse
|
44
|
Fang G, Lu H, Aboulkheyr Es H, Wang D, Liu Y, Warkiani ME, Lin G, Jin D. Unidirectional intercellular communication on a microfluidic chip. Biosens Bioelectron 2021; 175:112833. [PMID: 33288428 DOI: 10.1016/j.bios.2020.112833] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/29/2020] [Accepted: 11/17/2020] [Indexed: 12/15/2022]
Abstract
Cell co-culture serves as a standard method to study intercellular communication. However, random diffusion of signal molecules during co-culture may arouse crosstalk among different types of cells and hide directive signal-target responses. Here, a microfluidic chip is proposed to study unidirectional intercellular communication by spatially controlling the flow of the signal molecules. The chip contains two separated chambers connected by two channels where the culture media flows oppositely. A zigzag signal-blocking channel is designed to study the function of a specific signal. The chip is applied to study the unidirectional communication between tumor cells and stromal cells. It shows that the expression of α-smooth muscle actin (a marker of cancer-associated fibroblast (CAF)) of both MRC-5 fibroblasts and mesenchymal stem cells can be up-regulated only by the secreta from invasive MDA-MB-231 cells, but not from non-invasive MCF-7 cells. The proliferation of the tumor cells can be improved by the stromal cells. Moreover, transforming growth factor beta 1 is found as one of the main factors for CAF transformation via the signal-blocking function. The chip achieves unidirectional cell communication along X-axis, signal concentration gradient along Y-axis and 3D cell culture along Z-axis, which provides a useful tool for cell communication studies.
Collapse
Affiliation(s)
- Guocheng Fang
- Institute for Biomedical Materials and Devices, School of Mathematical and Physical Sciences, University of Technology Sydney, Broadway Ultimo, Sydney, NSW, 2007, Australia
| | - Hongxu Lu
- Institute for Biomedical Materials and Devices, School of Mathematical and Physical Sciences, University of Technology Sydney, Broadway Ultimo, Sydney, NSW, 2007, Australia.
| | - Hamidreza Aboulkheyr Es
- School of Biomedical Engineering, University of Technology Sydney, Broadway Ultimo, Sydney, NSW, 2007, Australia
| | - Dejiang Wang
- Institute for Biomedical Materials and Devices, School of Mathematical and Physical Sciences, University of Technology Sydney, Broadway Ultimo, Sydney, NSW, 2007, Australia
| | - Yuan Liu
- Institute for Biomedical Materials and Devices, School of Mathematical and Physical Sciences, University of Technology Sydney, Broadway Ultimo, Sydney, NSW, 2007, Australia
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Broadway Ultimo, Sydney, NSW, 2007, Australia
| | - Gungun Lin
- Institute for Biomedical Materials and Devices, School of Mathematical and Physical Sciences, University of Technology Sydney, Broadway Ultimo, Sydney, NSW, 2007, Australia
| | - Dayong Jin
- Institute for Biomedical Materials and Devices, School of Mathematical and Physical Sciences, University of Technology Sydney, Broadway Ultimo, Sydney, NSW, 2007, Australia; UTS-SUSTech Joint Research Centre for Biomedical Materials & Devices, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
45
|
Skala MC, Ayuso JM, Burkard ME, Deming DA. Breast cancer immunotherapy: current biomarkers and the potential of in vitro assays. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021; 21:100348. [PMID: 34901585 PMCID: PMC8654237 DOI: 10.1016/j.cobme.2021.100348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Breakthroughs in metastatic breast cancer care require new model systems that can identify the unique features and vulnerabilities of each cancer. Primary tumor cultures are proposed to efficiently screen multiple treatment options in a patient-specific strategy to maximize therapeutic benefit, minimize toxicity, and enable mechanistic insights that inspire future biomarkers for patient selection. To realize the potential of patient-specific cultures, new tools are needed to capture cell-by-cell variability in behavior and dynamic response to treatments in living 3D specimens. Potential bioengineering tools that can achieve this include optical microscopy to image single-cell dynamics and microphysiological in vitro systems to evaluate cell-cell interactions and immunotherapies.
Collapse
Affiliation(s)
- Melissa C. Skala
- Morgridge Institute for Research, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
| | - Jose M. Ayuso
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Mark E. Burkard
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
- Division of Hematology Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin, Madison, WI, USA
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin, Madison, WI, USA
| | - Dustin A. Deming
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
- Division of Hematology Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin, Madison, WI, USA
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
46
|
Shelton SE, Nguyen HT, Barbie DA, Kamm RD. Engineering approaches for studying immune-tumor cell interactions and immunotherapy. iScience 2021; 24:101985. [PMID: 33490895 PMCID: PMC7808917 DOI: 10.1016/j.isci.2020.101985] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
This review describes recent research that has advanced our understanding of the role of immune cells in the tumor microenvironment (TME) using advanced 3D in vitro models and engineering approaches. The TME can hinder effective eradication of tumor cells by the immune system, but immunotherapy has been able to reverse this effect in some cases. However, patient-to-patient variability in response suggests that we require deeper understanding of the mechanistic interactions between immune and tumor cells to improve response and develop novel therapeutics. Reconstruction of the TME using engineered 3D models allows high-resolution observation of cell interactions while allowing control of conditions such as hypoxia, matrix stiffness, and flow. Moreover, patient-derived organotypic models are an emerging tool for prediction of drug efficacy. This review highlights the importance of modeling and understanding the immune TME and describes new tools for identifying new biological targets, drug testing, and strategies for personalized medicine.
Collapse
Affiliation(s)
- Sarah E. Shelton
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Huu Tuan Nguyen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David A. Barbie
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Roger D. Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
47
|
Moses SR, Adorno JJ, Palmer AF, Song JW. Vessel-on-a-chip models for studying microvascular physiology, transport, and function in vitro. Am J Physiol Cell Physiol 2021; 320:C92-C105. [PMID: 33176110 PMCID: PMC7846973 DOI: 10.1152/ajpcell.00355.2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/20/2020] [Accepted: 11/08/2020] [Indexed: 12/15/2022]
Abstract
To understand how the microvasculature grows and remodels, researchers require reproducible systems that emulate the function of living tissue. Innovative contributions toward fulfilling this important need have been made by engineered microvessels assembled in vitro with microfabrication techniques. Microfabricated vessels, commonly referred to as "vessels-on-a-chip," are from a class of cell culture technologies that uniquely integrate microscale flow phenomena, tissue-level biomolecular transport, cell-cell interactions, and proper three-dimensional (3-D) extracellular matrix environments under well-defined culture conditions. Here, we discuss the enabling attributes of microfabricated vessels that make these models more physiological compared with established cell culture techniques and the potential of these models for advancing microvascular research. This review highlights the key features of microvascular transport and physiology, critically discusses the strengths and limitations of different microfabrication strategies for studying the microvasculature, and provides a perspective on current challenges and future opportunities for vessel-on-a-chip models.
Collapse
Affiliation(s)
- Savannah R Moses
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Jonathan J Adorno
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
| | - Andre F Palmer
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Jonathan W Song
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, Ohio
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| |
Collapse
|
48
|
Lee JJ, Berthier J, Kearney KE, Berthier E, Theberge AB. Open-Channel Capillary Trees and Capillary Pumping. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:12795-12803. [PMID: 32936651 PMCID: PMC8259885 DOI: 10.1021/acs.langmuir.0c01360] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Velocity of capillary flow in closed or open channels decreases as the flow proceeds down the length of the channel, varying as the inverse of the square root of time or as the inverse of travel distance. In order to increase the flow rate-and extend the duration of the flow-capillary pumps have been designed by mimicking the pumping principle of paper or cotton fibers. These designs provide a larger volume available for the wicking of the liquids. In microsystems for biotechnology, different designs have been developed based on experimental observation. In the present paper, the mechanisms at the basis of capillary pumping are investigated using a theoretical model for the flow in an open-channel "capillary tree" (i.e., an ensemble of channels with bifurcations mimicking the shape of a tree). The model is checked against experiments. Rules for obtaining better designs of capillary pumps are proposed; specifically, we find (1) when using a capillary tree with identical channel cross-sectional areas throughout, it is possible to maintain nearly constant flow rates throughout the channel network, (2) flow rate can be increased at each branch point of a capillary tree by slightly decreasing the areas of the channel cross section and decreasing the channel lengths at each level of ramification within the tree, and (3) higher order branching (trifurcations vs bifurcations) amplify the flow rate effect. This work lays the foundation for increasing the flow rate in open microfluidic channels driven by capillary flow; we expect this to have broad impact across open microfluidics for biological and chemical applications such as cell culture, sample preparation, separations, and on-chip reactions.
Collapse
Affiliation(s)
- Jing J. Lee
- Department of Chemistry, University of Washington, Box 351700, Seattle, Washington 98195, United States
| | - Jean Berthier
- Department of Chemistry, University of Washington, Box 351700, Seattle, Washington 98195, United States
| | - Kathleen E. Kearney
- Department of Chemistry, University of Washington, Box 351700, Seattle, Washington 98195, United States
| | - Erwin Berthier
- Department of Chemistry, University of Washington, Box 351700, Seattle, Washington 98195, United States
| | - Ashleigh B. Theberge
- Department of Chemistry, University of Washington, Box 351700, Seattle, Washington 98195, United States
- Department of Urology, University of Washington School of Medicine, Seattle, Washington 98105, United States
| |
Collapse
|
49
|
Heaster TM, Humayun M, Yu J, Beebe DJ, Skala MC. Autofluorescence Imaging of 3D Tumor-Macrophage Microscale Cultures Resolves Spatial and Temporal Dynamics of Macrophage Metabolism. Cancer Res 2020; 80:5408-5423. [PMID: 33093167 DOI: 10.1158/0008-5472.can-20-0831] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 07/17/2020] [Accepted: 10/19/2020] [Indexed: 12/24/2022]
Abstract
Macrophages within the tumor microenvironment (TME) exhibit a spectrum of protumor and antitumor functions, yet it is unclear how the TME regulates this macrophage heterogeneity. Standard methods to measure macrophage heterogeneity require destructive processing, limiting spatiotemporal studies of function within the live, intact 3D TME. Here, we demonstrate two-photon autofluorescence imaging of NAD(P)H and FAD to nondestructively resolve spatiotemporal metabolic heterogeneity of individual macrophages within 3D microscale TME models. Fluorescence lifetimes and intensities of NAD(P)H and FAD were acquired at 24, 48, and 72 hours poststimulation for mouse macrophages (RAW264.7) stimulated with IFNγ or IL4 plus IL13 in 2D culture, confirming that autofluorescence measurements capture known metabolic phenotypes. To quantify metabolic dynamics of macrophages within the TME, mouse macrophages or human monocytes (RAW264.7 or THP-1) were cultured alone or with breast cancer cells (mouse polyoma-middle T virus or primary human IDC) in 3D microfluidic platforms. Human monocytes and mouse macrophages in tumor cocultures exhibited significantly different FAD mean lifetimes and greater migration than monocultures at 24, 48, and 72 hours postseeding. In cocultures with primary human cancer cells, actively migrating monocyte-derived macrophages had greater redox ratios [NAD(P)H/FAD intensity] compared with passively migrating monocytes at 24 and 48 hours postseeding, reflecting metabolic heterogeneity in this subpopulation of monocytes. Genetic analyses further confirmed this metabolic heterogeneity. These results establish label-free autofluorescence imaging to quantify dynamic metabolism, polarization, and migration of macrophages at single-cell resolution within 3D microscale models. This combined culture and imaging system provides unique insights into spatiotemporal tumor-immune cross-talk within the 3D TME. SIGNIFICANCE: Label-free metabolic imaging and microscale culture technologies enable monitoring of single-cell macrophage metabolism, migration, and function in the 3D tumor microenvironment.
Collapse
Affiliation(s)
- Tiffany M Heaster
- Department of Biomedical Engineering, University of Wisconsin- Madison, Madison, Wisconsin.,Morgridge Institute for Research, Madison, Wisconsin
| | - Mouhita Humayun
- Department of Biomedical Engineering, University of Wisconsin- Madison, Madison, Wisconsin
| | - Jiaquan Yu
- Department of Biomedical Engineering, University of Wisconsin- Madison, Madison, Wisconsin.,Massachusetts Institute of Technology Koch Institute for Integrative Cancer Research, Cambridge, Massachusetts
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin- Madison, Madison, Wisconsin.,The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin.,Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, Wisconsin
| | - Melissa C Skala
- Department of Biomedical Engineering, University of Wisconsin- Madison, Madison, Wisconsin. .,Morgridge Institute for Research, Madison, Wisconsin.,The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
50
|
Berry SB, Haack AJ, Theberge AB, Brighenti S, Svensson M. Host and Pathogen Communication in the Respiratory Tract: Mechanisms and Models of a Complex Signaling Microenvironment. Front Med (Lausanne) 2020; 7:537. [PMID: 33015094 PMCID: PMC7511576 DOI: 10.3389/fmed.2020.00537] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 07/29/2020] [Indexed: 01/15/2023] Open
Abstract
Chronic lung diseases are a leading cause of morbidity and mortality across the globe, encompassing a diverse range of conditions from infections with pathogenic microorganisms to underlying genetic disorders. The respiratory tract represents an active interface with the external environment having the primary immune function of resisting pathogen intrusion and maintaining homeostasis in response to the myriad of stimuli encountered within its microenvironment. To perform these vital functions and prevent lung disorders, a chemical and biological cross-talk occurs in the complex milieu of the lung that mediates and regulates the numerous cellular processes contributing to lung health. In this review, we will focus on the role of cross-talk in chronic lung infections, and discuss how different cell types and signaling pathways contribute to the chronicity of infection(s) and prevent effective immune clearance of pathogens. In the lung microenvironment, pathogens have developed the capacity to evade mucosal immunity using different mechanisms or virulence factors, leading to colonization and infection of the host; such mechanisms include the release of soluble and volatile factors, as well as contact dependent (juxtracrine) interactions. We explore the diverse modes of communication between the host and pathogen in the lung tissue milieu in the context of chronic lung infections. Lastly, we review current methods and approaches used to model and study these host-pathogen interactions in vitro, and the role of these technological platforms in advancing our knowledge about chronic lung diseases.
Collapse
Affiliation(s)
- Samuel B. Berry
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Amanda J. Haack
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | | | - Susanna Brighenti
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Mattias Svensson
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|