1
|
Sadowski K, Jażdżewska A, Kozłowski J, Zacny A, Lorenc T, Olejarz W. Revolutionizing Glioblastoma Treatment: A Comprehensive Overview of Modern Therapeutic Approaches. Int J Mol Sci 2024; 25:5774. [PMID: 38891962 PMCID: PMC11172387 DOI: 10.3390/ijms25115774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 05/22/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
Glioblastoma is the most common malignant primary brain tumor in the adult population, with an average survival of 12.1 to 14.6 months. The standard treatment, combining surgery, radiotherapy, and chemotherapy, is not as efficient as we would like. However, the current possibilities are no longer limited to the standard therapies due to rapid advancements in biotechnology. New methods enable a more precise approach by targeting individual cells and antigens to overcome cancer. For the treatment of glioblastoma, these are gamma knife therapy, proton beam therapy, tumor-treating fields, EGFR and VEGF inhibitors, multiple RTKs inhibitors, and PI3K pathway inhibitors. In addition, the increasing understanding of the role of the immune system in tumorigenesis and the ability to identify tumor-specific antigens helped to develop immunotherapies targeting GBM and immune cells, including CAR-T, CAR-NK cells, dendritic cells, and immune checkpoint inhibitors. Each of the described methods has its advantages and disadvantages and faces problems, such as the inefficient crossing of the blood-brain barrier, various neurological and systemic side effects, and the escape mechanism of the tumor. This work aims to present the current modern treatments of glioblastoma.
Collapse
Affiliation(s)
- Karol Sadowski
- The Department of Histology and Embryology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland; (K.S.)
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Adrianna Jażdżewska
- The Department of Anatomy and Neurobiology, Medical University of Gdansk, Dębinki 1, 80-211 Gdansk, Poland;
| | - Jan Kozłowski
- The Department of Histology and Embryology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland; (K.S.)
| | - Aleksandra Zacny
- The Department of Histology and Embryology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland; (K.S.)
| | - Tomasz Lorenc
- Department of Radiology I, The Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland
| | - Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-091 Warsaw, Poland
| |
Collapse
|
2
|
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
|
3
|
Slika H, Karimov Z, Alimonti P, Abou-Mrad T, De Fazio E, Alomari S, Tyler B. Preclinical Models and Technologies in Glioblastoma Research: Evolution, Current State, and Future Avenues. Int J Mol Sci 2023; 24:16316. [PMID: 38003507 PMCID: PMC10671665 DOI: 10.3390/ijms242216316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Glioblastoma is the most common malignant primary central nervous system tumor and one of the most debilitating cancers. The prognosis of patients with glioblastoma remains poor, and the management of this tumor, both in its primary and recurrent forms, remains suboptimal. Despite the tremendous efforts that are being put forward by the research community to discover novel efficacious therapeutic agents and modalities, no major paradigm shifts have been established in the field in the last decade. However, this does not mirror the abundance of relevant findings and discoveries made in preclinical glioblastoma research. Hence, developing and utilizing appropriate preclinical models that faithfully recapitulate the characteristics and behavior of human glioblastoma is of utmost importance. Herein, we offer a holistic picture of the evolution of preclinical models of glioblastoma. We further elaborate on the commonly used in vitro and vivo models, delving into their development, favorable characteristics, shortcomings, and areas of potential improvement, which aids researchers in designing future experiments and utilizing the most suitable models. Additionally, this review explores progress in the fields of humanized and immunotolerant mouse models, genetically engineered animal models, 3D in vitro models, and microfluidics and highlights promising avenues for the future of preclinical glioblastoma research.
Collapse
Affiliation(s)
- Hasan Slika
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (H.S.); (Z.K.); (S.A.)
| | - Ziya Karimov
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (H.S.); (Z.K.); (S.A.)
- Faculty of Medicine, Ege University, 35100 Izmir, Turkey
| | - Paolo Alimonti
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy; (P.A.); (E.D.F.)
| | - Tatiana Abou-Mrad
- Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon;
- Department of Neurosurgery, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Emerson De Fazio
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy; (P.A.); (E.D.F.)
| | - Safwan Alomari
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (H.S.); (Z.K.); (S.A.)
| | - Betty Tyler
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (H.S.); (Z.K.); (S.A.)
| |
Collapse
|
4
|
Bouquerel C, Dubrova A, Hofer I, Phan DTT, Bernheim M, Ladaigue S, Cavaniol C, Maddalo D, Cabel L, Mechta-Grigoriou F, Wilhelm C, Zalcman G, Parrini MC, Descroix S. Bridging the gap between tumor-on-chip and clinics: a systematic review of 15 years of studies. LAB ON A CHIP 2023; 23:3906-3935. [PMID: 37592893 DOI: 10.1039/d3lc00531c] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
Over the past 15 years, the field of oncology research has witnessed significant progress in the development of new cell culture models, such as tumor-on-chip (ToC) systems. In this comprehensive overview, we present a multidisciplinary perspective by bringing together physicists, biologists, clinicians, and experts from pharmaceutical companies to highlight the current state of ToC research, its unique features, and the challenges it faces. To offer readers a clear and quantitative understanding of the ToC field, we conducted an extensive systematic analysis of more than 300 publications related to ToC from 2005 to 2022. ToC offer key advantages over other in vitro models by enabling precise control over various parameters. These parameters include the properties of the extracellular matrix, mechanical forces exerted on cells, the physico-chemical environment, cell composition, and the architecture of the tumor microenvironment. Such fine control allows ToC to closely replicate the complex microenvironment and interactions within tumors, facilitating the study of cancer progression and therapeutic responses in a highly representative manner. Importantly, by incorporating patient-derived cells or tumor xenografts, ToC models have demonstrated promising results in terms of clinical validation. We also examined the potential of ToC for pharmaceutical industries in which ToC adoption is expected to occur gradually. Looking ahead, given the high failure rate of clinical trials and the increasing emphasis on the 3Rs principles (replacement, reduction, refinement of animal experimentation), ToC models hold immense potential for cancer research. In the next decade, data generated from ToC models could potentially be employed for discovering new therapeutic targets, contributing to regulatory purposes, refining preclinical drug testing and reducing reliance on animal models.
Collapse
Affiliation(s)
- Charlotte Bouquerel
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
- Stress and Cancer Laboratory, Inserm, U830, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
- Fluigent, 67 avenue de Fontainebleau, 94270, Le Kremlin-Bicêtre, France
| | - Anastasiia Dubrova
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
| | - Isabella Hofer
- Stress and Cancer Laboratory, Inserm, U830, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
| | - Duc T T Phan
- Biomedicine Design, Pfizer Inc., San Diego, CA, USA
| | - Moencopi Bernheim
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
| | - Ségolène Ladaigue
- Stress and Cancer Laboratory, Inserm, U830, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
| | - Charles Cavaniol
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
| | - Danilo Maddalo
- Department of Translational Oncology, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Luc Cabel
- Institut Curie, Department of Medical Oncology, 26 rue d'Ulm, 75005, Paris, France
| | - Fatima Mechta-Grigoriou
- Stress and Cancer Laboratory, Inserm, U830, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
| | - Claire Wilhelm
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
| | - Gérard Zalcman
- Stress and Cancer Laboratory, Inserm, U830, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
- Université Paris Cité, Thoracic Oncology Department, INSERM CIC1425, Bichat Hospital, Cancer Institute AP-HP. Nord, Paris, France.
| | - Maria Carla Parrini
- Stress and Cancer Laboratory, Inserm, U830, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
| | - Stéphanie Descroix
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
| |
Collapse
|
5
|
Maurya R, Gohil N, Bhattacharjee G, Alzahrani KJ, Ramakrishna S, Singh V. Microfluidics for single cell analysis. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 186:203-215. [PMID: 35033285 DOI: 10.1016/bs.pmbts.2021.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Cells have several internal molecules that are present in low amounts and any fluctuation in its number drives a change in cell behavior. These molecules present inside the cells are continuously fluctuating, thus producing noises in the intrinsic environment and thereby directly affecting the cellular behavior. Single-cell analysis using microfluidics is an important tool for monitoring cell behavior by analyzing internal molecules. Several gene circuits have been designed for this purpose that are labeled with fluorescence encoding genes for monitoring cell dynamics and behavior. We discuss herewith designed and fabricated microfluidics devices that are used for trapping and tracking cells under controlled environmental conditions. This chapter highlights microfluidics chip for monitoring cells to promote their basic understanding.
Collapse
Affiliation(s)
- Rupesh Maurya
- Department of Biosciences, School of Science, Indrashil University, Rajpur, Mehsana, Gujarat, India
| | - Nisarg Gohil
- Department of Biosciences, School of Science, Indrashil University, Rajpur, Mehsana, Gujarat, India
| | - Gargi Bhattacharjee
- Department of Biosciences, School of Science, Indrashil University, Rajpur, Mehsana, Gujarat, India
| | - Khalid J Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea; College of Medicine, Hanyang University, Seoul, South Korea
| | - Vijai Singh
- Department of Biosciences, School of Science, Indrashil University, Rajpur, Mehsana, Gujarat, India.
| |
Collapse
|
6
|
Vázquez M, Anfossi L, Ben-Yoav H, Diéguez L, Karopka T, Della Ventura B, Abalde-Cela S, Minopoli A, Di Nardo F, Shukla VK, Teixeira A, Tvarijonaviciute A, Franco-Martínez L. Use of some cost-effective technologies for a routine clinical pathology laboratory. LAB ON A CHIP 2021; 21:4330-4351. [PMID: 34664599 DOI: 10.1039/d1lc00658d] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Classically, the need for highly sophisticated instruments with important economic costs has been a major limiting factor for clinical pathology laboratories, especially in developing countries. With the aim of making clinical pathology more accessible, a wide variety of free or economical technologies have been developed worldwide in the last few years. 3D printing and Arduino approaches can provide up to 94% economical savings in hardware and instrumentation in comparison to commercial alternatives. The vast selection of point-of-care-tests (POCT) currently available also limits the need for specific instruments or personnel, as they can be used almost anywhere and by anyone. Lastly, there are dozens of free and libre digital tools available in health informatics. This review provides an overview of the state-of-the-art on cost-effective alternatives with applications in routine clinical pathology laboratories. In this context, a variety of technologies including 3D printing and Arduino, lateral flow assays, plasmonic biosensors, and microfluidics, as well as laboratory information systems, are discussed. This review aims to serve as an introduction to different technologies that can make clinical pathology more accessible and, therefore, contribute to achieve universal health coverage.
Collapse
Affiliation(s)
- Mercedes Vázquez
- National Centre For Sensor Research, School of Chemical Sciences, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Laura Anfossi
- Department of Chemistry, University of Turin, Via Giuria, 5, I-10125 Turin, Italy
| | - Hadar Ben-Yoav
- Nanobioelectronics Laboratory (NBEL), Department of Biomedical Engineering, Ilse Katz Institute of Nanoscale Science and Technology, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Lorena Diéguez
- Medical Devices Research Group, International Iberian Nanotechnology Laboratory - INL, 4715-330 Braga, Portugal
| | | | - Bartolomeo Della Ventura
- Department of Physics "E. Pancini", University of Naples Federico II, Via Cintia 26, I-80126 Napoli, Italy
| | - Sara Abalde-Cela
- Medical Devices Research Group, International Iberian Nanotechnology Laboratory - INL, 4715-330 Braga, Portugal
| | - Antonio Minopoli
- Department of Physics "E. Pancini", University of Naples Federico II, Via Cintia 26, I-80126 Napoli, Italy
| | - Fabio Di Nardo
- Department of Chemistry, University of Turin, Via Giuria, 5, I-10125 Turin, Italy
| | - Vikas Kumar Shukla
- Nanobioelectronics Laboratory (NBEL), Department of Biomedical Engineering, Ilse Katz Institute of Nanoscale Science and Technology, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Alexandra Teixeira
- Medical Devices Research Group, International Iberian Nanotechnology Laboratory - INL, 4715-330 Braga, Portugal
| | - Asta Tvarijonaviciute
- Interdisciplinary Laboratory of Clinical Pathology, Interlab-UMU, Regional Campus of International Excellence 'Campus Mare Nostrum', University of Murcia, 30100 Murcia, Spain.
| | - Lorena Franco-Martínez
- Interdisciplinary Laboratory of Clinical Pathology, Interlab-UMU, Regional Campus of International Excellence 'Campus Mare Nostrum', University of Murcia, 30100 Murcia, Spain.
| |
Collapse
|
7
|
Jung Y, Son M, Nam YR, Choi J, Heath JR, Yang S. Microfluidic Single-Cell Proteomics Assay Chip: Lung Cancer Cell Line Case Study. MICROMACHINES 2021; 12:mi12101147. [PMID: 34683198 PMCID: PMC8541572 DOI: 10.3390/mi12101147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/16/2021] [Accepted: 09/20/2021] [Indexed: 12/22/2022]
Abstract
Cancer is a dynamic disease involving constant changes. With these changes, cancer cells become heterogeneous, resulting in varying sensitivity to chemotherapy. The heterogeneity of cancer cells plays a key role in chemotherapy resistance and cancer recurrence. Therefore, for effective treatment, cancer cells need to be analyzed at the single-cell level by monitoring various proteins and investigating their heterogeneity. We propose a microfluidic chip for a single-cell proteomics assay that is capable of analyzing complex cellular signaling systems to reveal the heterogeneity of cancer cells. The single-cell assay chip comprises (i) microchambers (n = 1376) for manipulating single cancer cells, (ii) micropumps for rapid single-cell lysis, and (iii) barcode immunosensors for detecting nine different secretory and intracellular proteins to reveal the correlation among cancer-related proteins. Using this chip, the single-cell proteomics of a lung cancer cell line, which may be easily masked in bulk analysis, were evaluated. By comparing changes in the level of protein secretion and heterogeneity in response to combinations of four anti-cancer drugs, this study suggests a new method for selecting the best combination of anti-cancer drugs. Subsequent preclinical and clinical trials should enable this platform to become applicable for patient-customized therapies.
Collapse
Affiliation(s)
- Yugyung Jung
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea; (Y.J.); (M.S.); (Y.R.N.)
| | - Minkook Son
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea; (Y.J.); (M.S.); (Y.R.N.)
| | - Yu Ri Nam
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea; (Y.J.); (M.S.); (Y.R.N.)
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Jongchan Choi
- School of Mechanical Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea;
- Institute for Systems Biology, Seattle, WA 98109, USA;
| | - James R. Heath
- Institute for Systems Biology, Seattle, WA 98109, USA;
- Department of Bioengineering, University of Washington, Seattle, WA 98105, USA
| | - Sung Yang
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea; (Y.J.); (M.S.); (Y.R.N.)
- School of Mechanical Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea;
- Correspondence:
| |
Collapse
|
8
|
Wen X, Takahashi S, Hatakeyama K, Kamei KI. Evaluation of the Effects of Solvents Used in the Fabrication of Microfluidic Devices on Cell Cultures. MICROMACHINES 2021; 12:550. [PMID: 34066183 PMCID: PMC8151832 DOI: 10.3390/mi12050550] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/08/2021] [Accepted: 05/10/2021] [Indexed: 12/15/2022]
Abstract
Microfluidic microphysiological systems (MPSs) or "organs-on-a-chip" are a promising alternative to animal models for drug screening and toxicology tests. However, most microfluidic devices employ polydimethylsiloxane (PDMS) as the structural material; and this has several drawbacks. Cyclo-olefin polymers (COPs) are more advantageous than PDMS and other thermoplastic materials because of their low drug absorption and autofluorescence. However, most COP-based microfluidic devices are fabricated by solvent bonding of the constituent parts. Notably, the remnant solvent can affect the cultured cells. This study employed a photobonding process with vacuum ultraviolet (VUV) light to fabricate microfluidic devices without using any solvent and compared their performance with that of solvent-bonded systems (using cyclohexane, dichloromethane, or toluene as the solvent) to investigate the effects of residual solvent on cell cultures. Quantitative immunofluorescence assays indicated that the coating efficiencies of extracellular matrix proteins (e.g., Matrigel and collagen I) were lower in solvent-bonded COP devices than those in VUV-bonded devices. Furthermore, the cytotoxicity of the systems was evaluated using SH-SY5Y neuroblastoma cells, and increased apoptosis was observed in the solvent-processed devices. These results provide insights into the effects of solvents used during the fabrication of microfluidic devices and can help prevent undesirable reactions and establish good manufacturing practices.
Collapse
Affiliation(s)
- Xiaopeng Wen
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku, Kyoto 606-8501, Japan;
| | - Seiichiro Takahashi
- Incubation Center Organs On Chip Project, Ushio INC, 1-6-5 Marunouchi, Chiyoda-ku, Tokyo 100-8150, Japan; (S.T.); (K.H.)
| | - Kenji Hatakeyama
- Incubation Center Organs On Chip Project, Ushio INC, 1-6-5 Marunouchi, Chiyoda-ku, Tokyo 100-8150, Japan; (S.T.); (K.H.)
| | - Ken-ichiro Kamei
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku, Kyoto 606-8501, Japan;
- Wuya College of Innovation, Shenyang Pharmaceutical University, Liaoning 110016, China
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Liaoning 110016, China
| |
Collapse
|
9
|
Lovett ML, Nieland TJ, Dingle YTL, Kaplan DL. Innovations in 3-Dimensional Tissue Models of Human Brain Physiology and Diseases. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1909146. [PMID: 34211358 PMCID: PMC8240470 DOI: 10.1002/adfm.201909146] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Indexed: 05/04/2023]
Abstract
3-dimensional (3D) laboratory tissue cultures have emerged as an alternative to traditional 2-dimensional (2D) culture systems that do not recapitulate native cell behavior. The discrepancy between in vivo and in vitro tissue-cell-molecular responses impedes understanding of human physiology in general and creates roadblocks for the discovery of therapeutic solutions. Two parallel approaches have emerged for the design of 3D culture systems. The first is biomedical engineering methodology, including bioengineered materials, bioprinting, microfluidics and bioreactors, used alone or in combination, to mimic the microenvironments of native tissues. The second approach is organoid technology, in which stem cells are exposed to chemical and/or biological cues to activate differentiation programs that are reminiscent of human (prenatal) development. This review article describes recent technological advances in engineering 3D cultures that more closely resemble the human brain. The contributions of in vitro 3D tissue culture systems to new insights in neurophysiology, neurological diseases and regenerative medicine are highlighted. Perspectives on designing improved tissue models of the human brain are offered, focusing on an integrative approach merging biomedical engineering tools with organoid biology.
Collapse
Affiliation(s)
- Michael L. Lovett
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155
| | - Thomas J.F. Nieland
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155
| | - Yu-Ting L. Dingle
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155
| |
Collapse
|
10
|
Labib M, Philpott DN, Wang Z, Nemr C, Chen JB, Sargent EH, Kelley SO. Magnetic Ranking Cytometry: Profiling Rare Cells at the Single-Cell Level. Acc Chem Res 2020; 53:1445-1457. [PMID: 32662263 DOI: 10.1021/acs.accounts.0c00179] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cellular heterogeneity in biological systems presents major challenges in the diagnosis and treatment of disease and also complicates the deconvolution of complex cellular phenomena. Single-cell analysis methods provide information that is not masked by the intrinsic heterogeneity of the bulk population and can therefore be applied to gain insights into heterogeneity among different cell subpopulations with fine resolution. Over the last 5 years, an explosion in the number of single-cell measurement methods has occurred. However, most of these methods are applicable to pure populations of cultured cells and are not able to handle high levels of phenotypic heterogeneity or a large background of nontarget cells. Microfluidics is an attractive tool for single cell manipulation as it enables individual encasing of single cells, allowing for high-throughput analysis with precise control of the local environment. Our laboratory has developed a new microfluidics-based analytical strategy to meet this unmet need referred to as magnetic ranking cytometry (MagRC). Cells expressing a biomarker of interest are labeled with receptor-coated magnetic nanoparticles and isolated from nontarget cells using a microfluidic device. The device ranks the cells according to the level of bound magnetic nanoparticles, which corresponds to the expression level of a target biomarker. Over the last several years, two generations of MagRC devices have been developed for different applications. The first-generation MagRC devices are powerful tools for the quantitation and analysis of rare cells present in heterogeneous samples, such as circulating tumor cells, stem cells, and pathogenic bacteria. The second-generation MagRC devices are compatible with the efficient recovery of cells sorted on the basis of protein expression and can be used to analyze large populations of cells and perform phenotypic CRISPR screens. To improve analytical precision, newer iterations of the first-generation and second-generation MagRC devices have been integrated with electrochemical sensors and Hall effect sensors, respectively. Both generations of MagRC devices permit the isolation of viable cells, which sets the stage for a wide range of applications, such as generating cell lines from rare cells and in vitro screening for effective therapeutic interventions in cancer patients to realize the promise of personalized medicine. This Account summarizes the development and application of the MagRC and describes a suite of advances that have enabled single-cell tumor cell analysis and monitoring tumor response to therapy, stem cell analysis, and detection of pathogens.
Collapse
Affiliation(s)
- Mahmoud Labib
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| | - David N. Philpott
- Department of Electrical & Computer Engineering, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Zongjie Wang
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G4, Canada
| | - Carine Nemr
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3G4, Canada
| | - Jenise B. Chen
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3G4, Canada
| | - Edward H. Sargent
- Department of Electrical & Computer Engineering, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Shana O. Kelley
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario M5S 3M2, Canada
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G4, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
11
|
Wu Z, Gong Z, Ao Z, Xu J, Cai H, Muhsen M, Heaps S, Bondesson M, Guo S, Guo F. Rapid Microfluidic Formation of Uniform Patient-Derived Breast Tumor Spheroids. ACS APPLIED BIO MATERIALS 2020; 3:6273-6283. [DOI: 10.1021/acsabm.0c00768] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Zhuhao Wu
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan 430072, China
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| | - Zhiyi Gong
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan 430072, China
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| | - Zheng Ao
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| | - Junhua Xu
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| | - Hongwei Cai
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| | - Maram Muhsen
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| | - Samuel Heaps
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| | - Maria Bondesson
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| | - Shishang Guo
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan 430072, China
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47405, United States
| |
Collapse
|
12
|
O’Donnell ST, Ross RP, Stanton C. The Progress of Multi-Omics Technologies: Determining Function in Lactic Acid Bacteria Using a Systems Level Approach. Front Microbiol 2020; 10:3084. [PMID: 32047482 PMCID: PMC6997344 DOI: 10.3389/fmicb.2019.03084] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022] Open
Abstract
Lactic Acid Bacteria (LAB) have long been recognized as having a significant impact ranging from commercial to health domains. A vast amount of research has been carried out on these microbes, deciphering many of the pathways and components responsible for these desirable effects. However, a large proportion of this functional information has been derived from a reductionist approach working with pure culture strains. This provides limited insight into understanding the impact of LAB within intricate systems such as the gut microbiome or multi strain starter cultures. Whole genome sequencing of strains and shotgun metagenomics of entire systems are powerful techniques that are currently widely used to decipher function in microbes, but they also have their limitations. An available genome or metagenome can provide an image of what a strain or microbiome, respectively, is potentially capable of and the functions that they may carry out. A top-down, multi-omics approach has the power to resolve the functional potential of an ecosystem into an image of what is being expressed, translated and produced. With this image, it is possible to see the real functions that members of a system are performing and allow more accurate and impactful predictions of the effects of these microorganisms. This review will discuss how technological advances have the potential to increase the yield of information from genomics, transcriptomics, proteomics and metabolomics. The potential for integrated omics to resolve the role of LAB in complex systems will also be assessed. Finally, the current software approaches for managing these omics data sets will be discussed.
Collapse
Affiliation(s)
- Shane Thomas O’Donnell
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
- Department of Microbiology, University College Cork – National University of Ireland, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - R. Paul Ross
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
- Department of Microbiology, University College Cork – National University of Ireland, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Catherine Stanton
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
- APC Microbiome Ireland, Cork, Ireland
| |
Collapse
|
13
|
Mofazzal Jahromi MA, Abdoli A, Rahmanian M, Bardania H, Bayandori M, Moosavi Basri SM, Kalbasi A, Aref AR, Karimi M, Hamblin MR. Microfluidic Brain-on-a-Chip: Perspectives for Mimicking Neural System Disorders. Mol Neurobiol 2019; 56:8489-8512. [PMID: 31264092 PMCID: PMC6842047 DOI: 10.1007/s12035-019-01653-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 05/15/2019] [Indexed: 01/09/2023]
Abstract
Neurodegenerative diseases (NDDs) include more than 600 types of nervous system disorders in humans that impact tens of millions of people worldwide. Estimates by the World Health Organization (WHO) suggest NDDs will increase by nearly 50% by 2030. Hence, development of advanced models for research on NDDs is needed to explore new therapeutic strategies and explore the pathogenesis of these disorders. Different approaches have been deployed in order to investigate nervous system disorders, including two-and three-dimensional (2D and 3D) cell cultures and animal models. However, these models have limitations, such as lacking cellular tension, fluid shear stress, and compression analysis; thus, studying the biochemical effects of therapeutic molecules on the biophysiological interactions of cells, tissues, and organs is problematic. The microfluidic "organ-on-a-chip" is an inexpensive and rapid analytical technology to create an effective tool for manipulation, monitoring, and assessment of cells, and investigating drug discovery, which enables the culture of various cells in a small amount of fluid (10-9 to 10-18 L). Thus, these chips have the ability to overcome the mentioned restrictions of 2D and 3D cell cultures, as well as animal models. Stem cells (SCs), particularly neural stem cells (NSCs), induced pluripotent stem cells (iPSCs), and embryonic stem cells (ESCs) have the capability to give rise to various neural system cells. Hence, microfluidic organ-on-a-chip and SCs can be used as potential research tools to study the treatment of central nervous system (CNS) and peripheral nervous system (PNS) disorders. Accordingly, in the present review, we discuss the latest progress in microfluidic brain-on-a-chip as a powerful and advanced technology that can be used in basic studies to investigate normal and abnormal functions of the nervous system.
Collapse
Affiliation(s)
- Mirza Ali Mofazzal Jahromi
- Department of Advanced Medical Sciences & Technologies, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
- Research Center for Noncommunicable Diseases, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Amir Abdoli
- Research Center for Noncommunicable Diseases, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
- Department of Parasitology and Mycology, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
- Zoonoses Research Center, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Mohammad Rahmanian
- Research Center for Noncommunicable Diseases, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
- Department of Anesthesiology, Critical Care, and Pain Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Hassan Bardania
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Mehrdad Bayandori
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Alireza Kalbasi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Amir Reza Aref
- Department of Cancer Biology, Center for Cancer Systems Biology, Dana-Farber Cancer Institute, Department of Genetics, Harvard Medical School, Boston, MA, 02215, USA
| | - Mahdi Karimi
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Research Center for Science and Technology in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Dermatology, Harvard Medical School, Boston, MA, USA.
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA.
| |
Collapse
|
14
|
FANG HS, LANG MF, SUN J. New Methods for Cell Cycle Analysis. CHINESE JOURNAL OF ANALYTICAL CHEMISTRY 2019. [DOI: 10.1016/s1872-2040(19)61186-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
15
|
Chen P, Chen D, Li S, Ou X, Liu BF. Microfluidics towards single cell resolution protein analysis. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2019.06.022] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
16
|
Zhang X, Li Y, Fang H, Wei H, Mu Y, Lang MF, Sun J. The influence of cell morphology on microfluidic single cell analysis. RSC Adv 2018; 9:139-144. [PMID: 35521600 PMCID: PMC9059331 DOI: 10.1039/c8ra08303g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 12/16/2018] [Indexed: 11/21/2022] Open
Abstract
Microfluidics has been widely used in single cell analysis. Current protocols allow either spread or round cells to be analyzed. However, the contribution of cell morphology to single cell analysis has not been noted. In this study, four proteins (EGFR, PTEN, pAKT, and pS6) in the EGFR signaling pathway are measured simultaneously using microfluidic image cytometry (MIC) in glioblastoma cells U87. The results show that the MIC technology can reveal different subsets of cells corresponding to the four protein expression levels no matter whether they are round or spread at the time of the measurements. However, sharper distinction is obtained from round cells, which implies that cellular heterogeneity can be better resolved with round cells during in situ protein quantification by imaging cytometry. This study calls attention to the role of cell morphology in single cell analysis. Future studies should examine whether differences in data interpretation resulting from cell morphology could reveal altered biological meanings.
Collapse
Affiliation(s)
- Xuxin Zhang
- Affiliated Zhongshan Hospital of Dalian University Dalian 116001 China
| | - Yanzhao Li
- Affiliated Zhongshan Hospital of Dalian University Dalian 116001 China
| | - Hanshu Fang
- Medical College, Institute of Microanalysis, Dalian University Dalian 116622 China
| | - Hongquan Wei
- First Hospital of China Medical University Shenyang 110001 China
| | - Ying Mu
- Institute of Cyber-Systems and Control, Zhejiang University Hangzhou 310007 China
| | - Ming-Fei Lang
- Medical College, Institute of Microanalysis, Dalian University Dalian 116622 China
| | - Jing Sun
- The Key Laboratory of Biomarker High-throughput Screening and Target Translation of Breast and Gastrointestinal Tumor of Liaoning Province, Dalian University Dalian 116622 China.,College of Environmental and Chemical Engineering, Institute of Microanalysis, Dalian University Dalian 116622 China
| |
Collapse
|
17
|
Wang Z, Zhang X. Single Cell Proteomics for Molecular Targets in Lung Cancer: High-Dimensional Data Acquisition and Analysis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1068:73-87. [PMID: 29943297 DOI: 10.1007/978-981-13-0502-3_7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the proteomic and genomic era, lung cancer researchers are increasingly under challenge with traditional protein analyzing tools. High output, multiplexed analytical procedures are in demand for disclosing the post-translational modification, molecular interactions and signaling pathways of proteins precisely, specifically, dynamically and systematically, as well as for identifying novel proteins and their functions. This could be better realized by single-cell proteomic methods than conventional proteomic methods. Using single-cell proteomic tools including flow cytometry, mass cytometry, microfluidics and chip technologies, chemical cytometry, single-cell western blotting, the quantity and functions of proteins are analyzed simultaneously. Aside from deciphering disease mechanisms, single-cell proteomic techniques facilitate the identification and screening of biomarkers, molecular targets and promising compounds as well. This review summarized single-cell proteomic tools and their use in lung cancer.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People's Hospital, Zhengzhou, China
| | - Xiaoju Zhang
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People's Hospital, Zhengzhou, China. .,Biomedical Research Center, Zhengzhou University People's Hospital, Zhengzhou, China.
| |
Collapse
|
18
|
Mashimo Y, Yoshioka M, Tokunaga Y, Fockenberg C, Terada S, Koyama Y, Shibata-Seki T, Yoshimoto K, Sakai R, Hakariya H, Liu L, Akaike T, Kobatake E, How SE, Uesugi M, Chen Y, Kamei KI. Fabrication of a Multiplexed Artificial Cellular MicroEnvironment Array. J Vis Exp 2018. [PMID: 30247461 DOI: 10.3791/57377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cellular microenvironments consist of a variety of cues, such as growth factors, extracellular matrices, and intercellular interactions. These cues are well orchestrated and are crucial in regulating cell functions in a living system. Although a number of researchers have attempted to investigate the correlation between environmental factors and desired cellular functions, much remains unknown. This is largely due to the lack of a proper methodology to mimic such environmental cues in vitro, and simultaneously test different environmental cues on cells. Here, we report an integrated platform of microfluidic channels and a nanofiber array, followed by high-content single-cell analysis, to examine stem cell phenotypes altered by distinct environmental factors. To demonstrate the application of this platform, this study focuses on the phenotypes of self-renewing human pluripotent stem cells (hPSCs). Here, we present the preparation procedures for a nanofiber array and the microfluidic structure in the fabrication of a Multiplexed Artificial Cellular MicroEnvironment (MACME) array. Moreover, overall steps of the single-cell profiling, cell staining with multiple fluorescent markers, multiple fluorescence imaging, and statistical analyses, are described.
Collapse
Affiliation(s)
- Yasumasa Mashimo
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University; Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology
| | - Momoko Yoshioka
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University
| | - Yumie Tokunaga
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University
| | | | - Shiho Terada
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University
| | - Yoshie Koyama
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University
| | - Teiko Shibata-Seki
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology
| | - Koki Yoshimoto
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University
| | - Risako Sakai
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University
| | - Hayase Hakariya
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University
| | - Li Liu
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University
| | - Toshihiro Akaike
- Biomaterials Center for Regenerative Medical Engineering, Foundation for Advancement of International Science
| | - Eiry Kobatake
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology
| | - Siew-Eng How
- Faculty of Science and Natural Resources, Universiti Malaysia Sabah
| | - Motonari Uesugi
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University; Institute for Chemical Research, Kyoto University
| | - Yong Chen
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University; Ecole Normale Supérieure
| | - Ken-Ichiro Kamei
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University;
| |
Collapse
|
19
|
Logun M, Zhao W, Mao L, Karumbaiah L. Microfluidics in Malignant Glioma Research and Precision Medicine. ADVANCED BIOSYSTEMS 2018; 2:1700221. [PMID: 29780878 PMCID: PMC5959050 DOI: 10.1002/adbi.201700221] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Indexed: 01/09/2023]
Abstract
Glioblastoma multiforme (GBM) is an aggressive form of brain cancer that has no effective treatments and a prognosis of only 12-15 months. Microfluidic technologies deliver microscale control of fluids and cells, and have aided cancer therapy as point-of-care devices for the diagnosis of breast and prostate cancers. However, a few microfluidic devices are developed to study malignant glioma. The ability of these platforms to accurately replicate the complex microenvironmental and extracellular conditions prevailing in the brain and facilitate the measurement of biological phenomena with high resolution and in a high-throughput manner could prove useful for studying glioma progression. These attributes, coupled with their relatively simple fabrication process, make them attractive for use as point-of-care diagnostic devices for detection and treatment of GBM. Here, the current issues that plague GBM research and treatment, as well as the current state of the art in glioma detection and therapy, are reviewed. Finally, opportunities are identified for implementing microfluidic technologies into research and diagnostics to facilitate the rapid detection and better therapeutic targeting of GBM.
Collapse
Affiliation(s)
- Meghan Logun
- Regenerative Bioscience Center, ADS Complex, University of Georgia, 425 River Road, Athens, GA 30602-2771, USA
| | - Wujun Zhao
- Department of Chemistry, University of Georgia, Athens, GA 30602-2771, USA
| | - Leidong Mao
- School of Electrical and Computer Engineering, College of Engineering, University of Georgia, Athens, GA 30602-2771, USA
| | - Lohitash Karumbaiah
- Regenerative Bioscience Center, ADS Complex, University of Georgia, 425 River Road, Athens, GA 30602-2771, USA
| |
Collapse
|
20
|
Yao N, Jan YJ, Cheng S, Chen JF, Chung LWK, Tseng HR, Posadas EM. Structure and function analysis in circulating tumor cells: using nanotechnology to study nuclear size in prostate cancer. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2018; 6:43-54. [PMID: 29666832 PMCID: PMC5902722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 03/19/2018] [Indexed: 06/08/2023]
Abstract
Professor Donald Coffey and his laboratory pioneered studies showing the relationships between nuclear shape and cellular function. In doing so, he and his students established the field of nuclear morphometry in prostate cancer. By using perioperative tissues via biopsies and surgical sampling, Dr. Coffey's team discovered that nuclear shape and other pathologic features correlated with clinical outcome measures. Cancer cells also exist outside of solid tumor masses as they can be shed from both primary and metastatic lesions into the circulatory system. The pool of these circulating tumor cells (CTCs) is heterogeneous. While some of these CTCs are passively shed into the circulation, others are active metastasizers with invasive potential. Advances in nanotechnology now make it possible to study morphologic features such as nuclear shape of CTCs in the bloodstream via liquid biopsy. Compared to traditional tissue sampling, liquid biopsy allows for minimally invasive, repetitive, and systemic disease sampling, which overcomes disease misrepresentation issues due to tumor temporospatial heterogeneity. Our team developed a novel liquid biopsy approach, the NanoVelcro assay, which allows us to identify morphologic heterogeneity in the CTC compartment. By applying classical methods of nuclear morphometry, we identified very small nuclear CTCs (vsnCTCs) in prostate cancer patients. Our initial studies showed that vsnCTCs strongly correlated with unfavorable clinical behaviors including the disposition to visceral metastases. These approaches may continue to yield additional insights into dynamic clinical behaviors, which creates an opportunity for more comprehensive and accurate cancer profiling. Ultimately, these advancements will allow physicians to employ more accurate and personalized treatments, helping the field reach the goal of true precision medicine.
Collapse
Affiliation(s)
- Nu Yao
- Urologic Oncology Program & Uro-Oncology Laboratories, Samuel Oschin Comprehensive Cancer InstituteLos Angeles, CA 90048, USA
| | - Yu-Jen Jan
- Urologic Oncology Program & Uro-Oncology Laboratories, Samuel Oschin Comprehensive Cancer InstituteLos Angeles, CA 90048, USA
| | - Shirley Cheng
- Urologic Oncology Program & Uro-Oncology Laboratories, Samuel Oschin Comprehensive Cancer InstituteLos Angeles, CA 90048, USA
| | - Jie-Fu Chen
- Urologic Oncology Program & Uro-Oncology Laboratories, Samuel Oschin Comprehensive Cancer InstituteLos Angeles, CA 90048, USA
| | - Leland WK Chung
- Cancer Biology Program, Samuel Oschin Comprehensive Cancer InstituteLos Angeles, CA 90048, USA
- Division Of Hematology/Oncology, Department of Medicine, Cedars-Sinai Medical CenterLos Angeles, CA, USA
| | - Hsian-Rong Tseng
- Department of Molecular and Medical Pharmacology, University of CaliforniaLos Angeles, USA
| | - Edwin M Posadas
- Urologic Oncology Program & Uro-Oncology Laboratories, Samuel Oschin Comprehensive Cancer InstituteLos Angeles, CA 90048, USA
- Translational Oncology Program, Samuel Oschin Comprehensive Cancer InstituteLos Angeles, CA 90048, USA
- Division Of Hematology/Oncology, Department of Medicine, Cedars-Sinai Medical CenterLos Angeles, CA, USA
| |
Collapse
|
21
|
Tsai HF, Trubelja A, Shen AQ, Bao G. Tumour-on-a-chip: microfluidic models of tumour morphology, growth and microenvironment. J R Soc Interface 2018. [PMID: 28637915 DOI: 10.1098/rsif.2017.0137] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cancer remains one of the leading causes of death, albeit enormous efforts to cure the disease. To overcome the major challenges in cancer therapy, we need to have a better understanding of the tumour microenvironment (TME), as well as a more effective means to screen anti-cancer drug leads; both can be achieved using advanced technologies, including the emerging tumour-on-a-chip technology. Here, we review the recent development of the tumour-on-a-chip technology, which integrates microfluidics, microfabrication, tissue engineering and biomaterials research, and offers new opportunities for building and applying functional three-dimensional in vitro human tumour models for oncology research, immunotherapy studies and drug screening. In particular, tumour-on-a-chip microdevices allow well-controlled microscopic studies of the interaction among tumour cells, immune cells and cells in the TME, of which simple tissue cultures and animal models are not amenable to do. The challenges in developing the next-generation tumour-on-a-chip technology are also discussed.
Collapse
Affiliation(s)
- Hsieh-Fu Tsai
- Micro/Bio/Nanofluidics Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa 904-0495, Japan
| | - Alen Trubelja
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Amy Q Shen
- Micro/Bio/Nanofluidics Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa 904-0495, Japan
| | - Gang Bao
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| |
Collapse
|
22
|
Jan YJ, Chen JF, Zhu Y, Lu YT, Chen SH, Chung H, Smalley M, Huang YW, Dong J, Chen LC, Yu HH, Tomlinson JS, Hou S, Agopian VG, Posadas EM, Tseng HR. NanoVelcro rare-cell assays for detection and characterization of circulating tumor cells. Adv Drug Deliv Rev 2018; 125:78-93. [PMID: 29551650 PMCID: PMC5993593 DOI: 10.1016/j.addr.2018.03.006] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 03/08/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022]
Abstract
Circulating tumor cells (CTCs) are cancer cells shredded from either a primary tumor or a metastatic site and circulate in the blood as the potential cellular origin of metastasis. By detecting and analyzing CTCs, we will be able to noninvasively monitor disease progression in individual cancer patients and obtain insightful information for assessing disease status, thus realizing the concept of "tumor liquid biopsy". However, it is technically challenging to identify CTCs in patient blood samples because of the extremely low abundance of CTCs among a large number of hematologic cells. In order to address this challenge, our research team at UCLA pioneered a unique concept of "NanoVelcro" cell-affinity substrates, in which CTC capture agent-coated nanostructured substrates were utilized to immobilize CTCs with remarkable efficiency. Four generations of NanoVelcro CTC assays have been developed over the past decade for a variety of clinical utilities. The 1st-gen NanoVelcro Chips, composed of a silicon nanowire substrate (SiNS) and an overlaid microfluidic chaotic mixer, were created for CTC enumeration. The 2nd-gen NanoVelcro Chips (i.e., NanoVelcro-LMD), based on polymer nanosubstrates, were developed for single-CTC isolation in conjunction with the use of the laser microdissection (LMD) technique. By grafting thermoresponsive polymer brushes onto SiNS, the 3rd-gen Thermoresponsive NanoVelcro Chips have demonstrated the capture and release of CTCs at 37 and 4 °C respectively, thereby allowing for rapid CTC purification while maintaining cell viability and molecular integrity. Fabricated with boronic acid-grafted conducting polymer-based nanomaterial on chip surface, the 4th-gen NanoVelcro Chips (Sweet chip) were able to purify CTCs with well-preserved RNA transcripts, which could be used for downstream analysis of several cancer specific RNA biomarkers. In this review article, we will summarize the development of the four generations of NanoVelcro CTC assays, and the clinical applications of each generation of devices.
Collapse
Affiliation(s)
- Yu Jen Jan
- Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jie-Fu Chen
- Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yazhen Zhu
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yi-Tsung Lu
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Szu Hao Chen
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Howard Chung
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Matthew Smalley
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA; CytoLumina Technologies Corp., Los Angeles, CA, USA
| | - Yen-Wen Huang
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA; CytoLumina Technologies Corp., Los Angeles, CA, USA
| | - Jiantong Dong
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Li-Ching Chen
- Department of Obstetrics and Gynecology, Cathay General Hospital, Taipei, Taiwan
| | - Hsiao-Hua Yu
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - James S Tomlinson
- Department of Surgery, University of California, Los Angeles, Los Angeles, CA, USA; Center for Pancreatic Disease, University of California, Los Angeles, Los Angeles, CA, USA; Department of Surgery, Greater Los Angeles Veteran's Affairs Administration, Los Angeles, CA, USA
| | - Shuang Hou
- Department of Surgery, University of California, Los Angeles, Los Angeles, CA, USA
| | - Vatche G Agopian
- Department of Surgery, University of California, Los Angeles, Los Angeles, CA, USA; Liver Transplantation and Hepatobiliary Surgery, University of California, Los Angeles, Los Angeles, CA, USA
| | - Edwin M Posadas
- Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hsian-Rong Tseng
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
23
|
Abstract
Cancer immunotherapy fights against cancer by modulating the immune response and is delivering encouraging results in clinical treatments. However, it is challenging to achieve durable response in all cancer patients during treatment due to the diversity and dynamic nature of immune system as well as inter- and intratumor heterogeneity. A comprehensive assessment of system immunity and tumor microenvironment is crucial for effective and safe cancer therapy, which can potentially be resolved by single-cell proteomic analysis. Single-cell proteomic technologies enable system-wide profiling of protein levels in a number of single cells within the immune system and tumor microenvironment, and thereby provide direct assessment of the functional state of the immune cells and tumor-immune interaction that could be used to evaluate efficacy of immunotherapy and to improve clinical outcome. In this chapter, we summarized current single-cell proteomic technologies and their applications in cancer immunotherapy.
Collapse
|
24
|
Murphy TW, Zhang Q, Naler LB, Ma S, Lu C. Recent advances in the use of microfluidic technologies for single cell analysis. Analyst 2017; 143:60-80. [PMID: 29170786 PMCID: PMC5839671 DOI: 10.1039/c7an01346a] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The inherent heterogeneity in cell populations has become of great interest and importance as analytical techniques have improved over the past decades. With the advent of personalized medicine, understanding the impact of this heterogeneity has become an important challenge for the research community. Many different microfluidic approaches with varying levels of throughput and resolution exist to study single cell activity. In this review, we take a broad view of the recent microfluidic developments in single cell analysis based on microwell, microchamber, and droplet platforms. We cover physical, chemical, and molecular biology approaches for cellular and molecular analysis including newly emerging genome-wide analysis.
Collapse
Affiliation(s)
- Travis W Murphy
- Department of Chemical Engineering, Virginia Tech, Blacksburg, VA 24061, USA.
| | | | | | | | | |
Collapse
|
25
|
Hou S, Chen JF, Song M, Zhu Y, Jan YJ, Chen SH, Weng TH, Ling DA, Chen SF, Ro T, Liang AJ, Lee T, Jin H, Li M, Liu L, Hsiao YS, Chen P, Yu HH, Tsai MS, Pisarska MD, Chen A, Chen LC, Tseng HR. Imprinted NanoVelcro Microchips for Isolation and Characterization of Circulating Fetal Trophoblasts: Toward Noninvasive Prenatal Diagnostics. ACS NANO 2017; 11:8167-8177. [PMID: 28721719 PMCID: PMC5614709 DOI: 10.1021/acsnano.7b03073] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Circulating fetal nucleated cells (CFNCs) in maternal blood offer an ideal source of fetal genomic DNA for noninvasive prenatal diagnostics (NIPD). We developed a class of nanoVelcro microchips to effectively enrich a subcategory of CFNCs, i.e., circulating trophoblasts (cTBs) from maternal blood, which can then be isolated with single-cell resolution by a laser capture microdissection (LCM) technique for downstream genetic testing. We first established a nanoimprinting fabrication process to prepare the LCM-compatible nanoVelcro substrates. Using an optimized cTB-capture condition and an immunocytochemistry protocol, we were able to identify and isolate single cTBs (Hoechst+/CK7+/HLA-G+/CD45-, 20 μm > sizes > 12 μm) on the imprinted nanoVelcro microchips. Three cTBs were polled to ensure reproducible whole genome amplification on the cTB-derived DNA, paving the way for cTB-based array comparative genomic hybridization (aCGH) and short tandem repeats analysis. Using maternal blood samples collected from expectant mothers carrying a single fetus, the cTB-derived aCGH data were able to detect fetal genders and chromosomal aberrations, which had been confirmed by standard clinical practice. Our results support the use of nanoVelcro microchips for cTB-based noninvasive prenatal genetic testing, which holds potential for further development toward future NIPD solution.
Collapse
Affiliation(s)
- Shuang Hou
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095-1770, United States
| | - Jie-Fu Chen
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095-1770, United States
| | - Min Song
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095-1770, United States
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095-1770, United States
- Department of Pathology, Guangdong Provincial Hospital of TCM, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yu Jen Jan
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095-1770, United States
| | - Szu Hao Chen
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095-1770, United States
| | - Tzu-Hua Weng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095-1770, United States
| | - Dean-An Ling
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095-1770, United States
| | - Shang-Fu Chen
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095-1770, United States
| | - Tracy Ro
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095-1770, United States
| | - An-Jou Liang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095-1770, United States
| | - Tom Lee
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095-1770, United States
| | - Helen Jin
- PacGenomics, Agoura Hills, California 91301, United States
| | - Man Li
- PacGenomics, Agoura Hills, California 91301, United States
| | - Lian Liu
- PacGenomics, Agoura Hills, California 91301, United States
| | - Yu-Sheng Hsiao
- Research Center for Applied Sciences, Taipei, Taiwan, 115
| | - Peilin Chen
- Research Center for Applied Sciences, Taipei, Taiwan, 115
| | - Hsiao-Hua Yu
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan, 115
| | - Ming-Song Tsai
- Department of Obstetrics and Gynecology, Cathay General Hospital, Taipei, Taiwan 106
| | - Margareta D. Pisarska
- Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Angela Chen
- Department of Obstetrics and Gynecology, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Li-Ching Chen
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095-1770, United States
- Department of Obstetrics and Gynecology, Cathay General Hospital, Taipei, Taiwan 106
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095-1770, United States
| |
Collapse
|
26
|
Yu ZTF, Joseph JG, Liu SX, Cheung MK, Haffey PJ, Kurabayashi K, Fu J. Centrifugal microfluidics for sorting immune cells from whole blood. SENSORS AND ACTUATORS. B, CHEMICAL 2017; 245:1050-1061. [PMID: 28966475 PMCID: PMC5619665 DOI: 10.1016/j.snb.2017.01.113] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Sorting and enumeration of immune cells from blood are critical operations involved in many clinical applications. Conventional methods for sorting and counting immune cells from blood, such as flow cytometry and hemocytometers, are tedious, inaccurate, and difficult for implementation for point-of-care (POC) testing. Herein we developed a microscale centrifugal technology termed Centrifugal Microfluidic Chip (CMC) capable of sorting immune cells from blood and in situ cellular analysis in a laboratory setting. Operation of the CMC entailed a blood specimen layered on a density gradient medium and centrifuged in microfluidic channels where immune cell subpopulations could rapidly be sorted into distinct layers according to their density differentials. We systematically studied effects of different blocking molecules for surface passivation of the CMC. We further demonstrated the applicability of CMCs for rapid separation of minimally processed human whole blood without affecting immune cell viability. Multi-color imaging and analysis of immune cell distributions and enrichment such as recovery and purity rates of peripheral blood mononuclear cells (PBMCs) were demonstrated using CMCs. Given its design and operation simplicity, portability, blood cell sorting efficiency, and in situ cellular analysis capability, the CMC holds promise for blood-based diagnosis and disease monitoring in POC applications.
Collapse
Affiliation(s)
- Zeta Tak For Yu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Jophin George Joseph
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Chemical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana, India 502285
| | - Shirley Xiaosu Liu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Mei Ki Cheung
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Parker James Haffey
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Katsuo Kurabayashi
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Michigan Center for Integrative Research in Critical Care, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
27
|
Lu Y, Yang L, Wei W, Shi Q. Microchip-based single-cell functional proteomics for biomedical applications. LAB ON A CHIP 2017; 17:1250-1263. [PMID: 28280819 PMCID: PMC5459479 DOI: 10.1039/c7lc00037e] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Cellular heterogeneity has been widely recognized but only recently have single cell tools become available that allow characterizing heterogeneity at the genomic and proteomic levels. We review the technological advances in microchip-based toolkits for single-cell functional proteomics. Each of these tools has distinct advantages and limitations, and a few have advanced toward being applied to address biological or clinical problems that traditional population-based methods fail to address. High-throughput single-cell proteomic assays generate high-dimensional data sets that contain new information and thus require developing new analytical frameworks to extract new biology. In this review article, we highlight a few biological and clinical applications in which microchip-based single-cell proteomic tools provide unique advantages. The examples include resolving functional heterogeneity and dynamics of immune cells, dissecting cell-cell interaction by creating a well-controlled on-chip microenvironment, capturing high-resolution snapshots of immune system functions in patients for better immunotherapy and elucidating phosphoprotein signaling networks in cancer cells for guiding effective molecularly targeted therapies.
Collapse
Affiliation(s)
- Yao Lu
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Liu Yang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Wei Wei
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA. and Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Qihui Shi
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
28
|
Abstract
Single-cell cell cycle analysis is an emerging technique that requires detailed exploration of the image analysis process. In this study, we established a microfluidic single-cell cell cycle analysis method that can analyze cells in small numbers and in situ on a microfluidic chip. In addition, factors that influenced the analysis were carefully investigated. U87 or HeLa cells were seeded and attached to microfluidic channels before measurement. Cell nucleic DNA was imaged by 4′-6-diamidino-2-phenylindole (DAPI) staining under a fluorescent microscope and subsequently fluorescent intensities of the cell nuclei DNA were converted to depict histograms for cell cycle phases. DAPI concentration, microscopic magnification, exposure time and cell number were examined for optimal cell cycle analysis conditions. The results showed that as few as a few hundred cells could be measured by DAPI staining in the range of 0.4–0.6 μg/mL to depict histograms with typical cell cycle phase distribution. Microscopic magnification during image acquisition, however, could distort the phase distribution. Exposure time did not significantly affect the cell cycle analysis. Furthermore, cell cycle inhibitor rapamycin treatment changed the cell cycle phase distribution as expected. In conclusion, a method for microfluidic single-cell cell cycle analysis of spread cells in situ was developed. Factors such as dye concentration and microscopic magnification had more influence on cell cycle phase distribution. Further studies will focus on detail differentiation of cell cycle phases and the application of such a method for biological meanings.
Collapse
|
29
|
Khoo BL, Chaudhuri PK, Lim CT, Warkiani ME. Advancing Techniques and Insights in Circulating Tumor Cell (CTC) Research. CANCER DRUG DISCOVERY AND DEVELOPMENT 2017:71-94. [DOI: 10.1007/978-3-319-45397-2_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
30
|
Tumour Heterogeneity: The Key Advantages of Single-Cell Analysis. Int J Mol Sci 2016; 17:ijms17122142. [PMID: 27999407 PMCID: PMC5187942 DOI: 10.3390/ijms17122142] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/12/2016] [Accepted: 12/13/2016] [Indexed: 01/06/2023] Open
Abstract
Tumour heterogeneity refers to the fact that different tumour cells can show distinct morphological and phenotypic profiles, including cellular morphology, gene expression, metabolism, motility, proliferation and metastatic potential. This phenomenon occurs both between tumours (inter-tumour heterogeneity) and within tumours (intra-tumour heterogeneity), and it is caused by genetic and non-genetic factors. The heterogeneity of cancer cells introduces significant challenges in using molecular prognostic markers as well as for classifying patients that might benefit from specific therapies. Thus, research efforts for characterizing heterogeneity would be useful for a better understanding of the causes and progression of disease. It has been suggested that the study of heterogeneity within Circulating Tumour Cells (CTCs) could also reflect the full spectrum of mutations of the disease more accurately than a single biopsy of a primary or metastatic tumour. In previous years, many high throughput methodologies have raised for the study of heterogeneity at different levels (i.e., RNA, DNA, protein and epigenetic events). The aim of the current review is to stress clinical implications of tumour heterogeneity, as well as current available methodologies for their study, paying specific attention to those able to assess heterogeneity at the single cell level.
Collapse
|
31
|
Kashaninejad N, Nikmaneshi MR, Moghadas H, Kiyoumarsi Oskouei A, Rismanian M, Barisam M, Saidi MS, Firoozabadi B. Organ-Tumor-on-a-Chip for Chemosensitivity Assay: A Critical Review. MICROMACHINES 2016; 7:mi7080130. [PMID: 30404302 PMCID: PMC6190381 DOI: 10.3390/mi7080130] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 06/22/2016] [Accepted: 07/18/2016] [Indexed: 01/09/2023]
Abstract
With a mortality rate over 580,000 per year, cancer is still one of the leading causes of death worldwide. However, the emerging field of microfluidics can potentially shed light on this puzzling disease. Unique characteristics of microfluidic chips (also known as micro-total analysis system) make them excellent candidates for biological applications. The ex vivo approach of tumor-on-a-chip is becoming an indispensable part of personalized medicine and can replace in vivo animal testing as well as conventional in vitro methods. In tumor-on-a-chip, the complex three-dimensional (3D) nature of malignant tumor is co-cultured on a microfluidic chip and high throughput screening tools to evaluate the efficacy of anticancer drugs are integrated on the same chip. In this article, we critically review the cutting edge advances in this field and mainly categorize each tumor-on-a-chip work based on its primary organ. Specifically, design, fabrication and characterization of tumor microenvironment; cell culture technique; transferring mechanism of cultured cells into the microchip; concentration gradient generators for drug delivery; in vitro screening assays of drug efficacy; and pros and cons of each microfluidic platform used in the recent literature will be discussed separately for the tumor of following organs: (1) Lung; (2) Bone marrow; (3) Brain; (4) Breast; (5) Urinary system (kidney, bladder and prostate); (6) Intestine; and (7) Liver. By comparing these microchips, we intend to demonstrate the unique design considerations of each tumor-on-a-chip based on primary organ, e.g., how microfluidic platform of lung-tumor-on-a-chip may differ from liver-tumor-on-a-chip. In addition, the importance of heart–liver–intestine co-culture with microvasculature in tumor-on-a-chip devices for in vitro chemosensitivity assay will be discussed. Such system would be able to completely evaluate the absorption, distribution, metabolism, excretion and toxicity (ADMET) of anticancer drugs and more realistically recapitulate tumor in vivo-like microenvironment.
Collapse
Affiliation(s)
- Navid Kashaninejad
- School of Mechanical Engineering, Sharif University of Technology, 11155-9567 Tehran, Iran.
| | | | - Hajar Moghadas
- School of Mechanical Engineering, Sharif University of Technology, 11155-9567 Tehran, Iran.
| | | | - Milad Rismanian
- School of Mechanical Engineering, Sharif University of Technology, 11155-9567 Tehran, Iran.
| | - Maryam Barisam
- School of Mechanical Engineering, Sharif University of Technology, 11155-9567 Tehran, Iran.
| | - Mohammad Said Saidi
- School of Mechanical Engineering, Sharif University of Technology, 11155-9567 Tehran, Iran.
| | - Bahar Firoozabadi
- School of Mechanical Engineering, Sharif University of Technology, 11155-9567 Tehran, Iran.
| |
Collapse
|
32
|
Chen JF, Zhu Y, Lu YT, Hodara E, Hou S, Agopian VG, Tomlinson JS, Posadas EM, Tseng HR. Clinical Applications of NanoVelcro Rare-Cell Assays for Detection and Characterization of Circulating Tumor Cells. Theranostics 2016; 6:1425-39. [PMID: 27375790 PMCID: PMC4924510 DOI: 10.7150/thno.15359] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 05/06/2016] [Indexed: 12/22/2022] Open
Abstract
Liquid biopsy of tumor through isolation of circulating tumor cells (CTCs) allows non-invasive, repetitive, and systemic sampling of disease. Although detecting and enumerating CTCs is of prognostic significance in metastatic cancer, it is conceivable that performing molecular and functional characterization on CTCs will reveal unprecedented insight into the pathogenic mechanisms driving lethal disease. Nanomaterial-embedded cancer diagnostic platforms, i.e., NanoVelcro CTC Assays represent a unique rare-cell sorting method that enables detection isolation, and characterization of CTCs in peripheral blood, providing an opportunity to noninvasively monitor disease progression in individual cancer patients. Over the past decade, a series of NanoVelcro CTC Assays has been demonstrated for exploring the full potential of CTCs as a clinical biomarker, including CTC enumeration, phenotyping, genotyping and expression profiling. In this review article, the authors will briefly introduce the development of three generations of NanoVelcro CTC Assays, and highlight the clinical applications of each generation for various types of solid cancers, including prostate cancer, pancreatic cancer, lung cancer, and melanoma.
Collapse
Affiliation(s)
- Jie-Fu Chen
- 1. Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Yazhen Zhu
- 2. Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, California, USA;; 3. Department of Pathology, Guangdong Provincial Hospital of TCM, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yi-Tsung Lu
- 1. Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Elisabeth Hodara
- 1. Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Shuang Hou
- 3. Department of Pathology, Guangdong Provincial Hospital of TCM, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Vatche G Agopian
- 4. Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA;; 5. Liver Transplantation and Hepatobiliary Surgery, University of California, Los Angeles, Los Angeles, California, USA
| | - James S Tomlinson
- 4. Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA;; 6. Center for Pancreatic Disease, University of California, Los Angeles, Los Angeles, California, USA;; 7. Department of Surgery Greater Los Angeles Veteran's Affairs Administration, Los Angeles, California, USA
| | - Edwin M Posadas
- 1. Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Hsian-Rong Tseng
- 2. Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
33
|
Zhang Y, Tang Y, Sun S, Wang Z, Wu W, Zhao X, Czajkowsky DM, Li Y, Tian J, Xu L, Wei W, Deng Y, Shi Q. Single-cell codetection of metabolic activity, intracellular functional proteins, and genetic mutations from rare circulating tumor cells. Anal Chem 2016; 87:9761-8. [PMID: 26378744 DOI: 10.1021/acs.analchem.5b01901] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The high glucose uptake and activation of oncogenic signaling pathways in cancer cells has long made these features, together with the mutational spectrum, prime diagnostic targets of circulating tumor cells (CTCs). Further, an ability to characterize these properties at a single cell resolution is widely believed to be essential, as the known extensive heterogeneity in CTCs can obscure important correlations in data obtained from cell population-based methods. However, to date, it has not been possible to quantitatively measure metabolic, proteomic, and genetic data from a single CTC. Here we report a microchip-based approach that allows for the codetection of glucose uptake, intracellular functional proteins, and genetic mutations at the single-cell level from rare tumor cells. The microchip contains thousands of nanoliter grooves (nanowells) that isolate individual CTCs and allow for the assessment of their glucose uptake via imaging of a fluorescent glucose analog, quantification of a panel of intracellular signaling proteins using a miniaturized antibody barcode microarray, and retrieval of the individual cell nuclei for subsequent off-chip genome amplification and sequencing. This approach integrates molecular-scale information on the metabolic, proteomic, and genetic status of single cells and permits the inference of associations between genetic signatures, energy consumption, and phosphoproteins oncogenic signaling activities in CTCs isolated from blood samples of patients. Importantly, this microchip chip-based approach achieves this multidimensional molecular analysis with minimal cell loss (<20%), which is the bottleneck of the rare cell analysis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yan Li
- Shanghai Municipal Hospital of Traditional Chinese Medicine , Shanghai 200071, P.R. China
| | - Jianhui Tian
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine , Shanghai 200032, P.R. China
| | - Ling Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine , Shanghai 200032, P.R. China
| | - Wei Wei
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California , Los Angeles, California 90095, United States
| | | | | |
Collapse
|
34
|
Konry T, Sarkar S, Sabhachandani P, Cohen N. Innovative Tools and Technology for Analysis of Single Cells and Cell-Cell Interaction. Annu Rev Biomed Eng 2016; 18:259-84. [PMID: 26928209 DOI: 10.1146/annurev-bioeng-090215-112735] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Heterogeneity in single-cell responses and intercellular interactions results from complex regulation of cell-intrinsic and environmental factors. Single-cell analysis allows not only detection of individual cellular characteristics but also correlation of genetic content with phenotypic traits in the same cell. Technological advances in micro- and nanofabrication have benefited single-cell analysis by allowing precise control of the localized microenvironment, cell manipulation, and sensitive detection capabilities. Additionally, microscale techniques permit rapid, high-throughput, multiparametric screening that has become essential for -omics research. This review highlights innovative applications of microscale platforms in genetic, proteomic, and metabolic detection in single cells; cell sorting strategies; and heterotypic cell-cell interaction. We discuss key design aspects of single-cell localization and isolation in microfluidic systems, dynamic and endpoint analyses, and approaches that integrate highly multiplexed detection of various intracellular species.
Collapse
Affiliation(s)
- Tania Konry
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts 02115; , , ,
| | - Saheli Sarkar
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts 02115; , , ,
| | - Pooja Sabhachandani
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts 02115; , , ,
| | - Noa Cohen
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts 02115; , , ,
| |
Collapse
|
35
|
Khoo BL, Chaudhuri PK, Ramalingam N, Tan DSW, Lim CT, Warkiani ME. Single-cell profiling approaches to probing tumor heterogeneity. Int J Cancer 2016; 139:243-55. [PMID: 26789729 DOI: 10.1002/ijc.30006] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 12/10/2015] [Accepted: 01/08/2016] [Indexed: 01/08/2023]
Abstract
Tumor heterogeneity is a major hindrance in cancer classification, diagnosis and treatment. Recent technological advances have begun to reveal the true extent of its heterogeneity. Single-cell analysis (SCA) is emerging as an important approach to detect variations in morphology, genetic or proteomic expression. In this review, we revisit the issue of inter- and intra-tumor heterogeneity, and list various modes of SCA techniques (cell-based, nucleic acid-based, protein-based, metabolite-based and lipid-based) presently used for cancer characterization. We further discuss the advantages of SCA over pooled cell analysis, as well as the limitations of conventional techniques. Emerging trends, such as high-throughput sequencing, are also mentioned as improved means for cancer profiling. Collectively, these applications have the potential for breakthroughs in cancer treatment.
Collapse
Affiliation(s)
- Bee Luan Khoo
- Mechanobiology Institute, National University of Singapore.,BioSystems and Micromechanics (BioSyM) IRG, Singapore-MIT Alliance for Research and Technology (SMART) Centre, Singapore
| | | | | | - Daniel Shao Weng Tan
- Division of Medical Oncology, National Cancer Centre Singapore.,Cancer Stem Cell Biology, Genome Institute of Singapore
| | - Chwee Teck Lim
- Mechanobiology Institute, National University of Singapore.,BioSystems and Micromechanics (BioSyM) IRG, Singapore-MIT Alliance for Research and Technology (SMART) Centre, Singapore.,Department of Biomedical Engineering, National University of Singapore
| | - Majid Ebrahimi Warkiani
- BioSystems and Micromechanics (BioSyM) IRG, Singapore-MIT Alliance for Research and Technology (SMART) Centre, Singapore.,School of Mechanical and Manufacturing Engineering, Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW, 2052, Australia
| |
Collapse
|
36
|
Liu Y, Du J, Choi JS, Chen KJ, Hou S, Yan M, Lin WY, Chen KS, Ro T, Lipshutz GS, Wu L, Shi L, Lu Y, Tseng HR, Wang H. A High-Throughput Platform for Formulating and Screening Multifunctional Nanoparticles Capable of Simultaneous Delivery of Genes and Transcription Factors. Angew Chem Int Ed Engl 2016; 55:169-73. [PMID: 26768819 PMCID: PMC5577986 DOI: 10.1002/anie.201507546] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 10/21/2015] [Indexed: 01/09/2023]
Abstract
Simultaneous delivery of multiple genes and proteins (e.g., transcription factors; TFs) is an emerging issue surrounding therapeutic research due to their ability to regulate cellular circuitry. Current gene and protein delivery strategies, however, are based on slow batch synthesis, which is ineffective, poorly controlled, and incapable of simultaneous delivery of both genes and proteins with synergistic functions. Consequently, advances in this field have been limited to in vitro studies. Here, by integrating microfluidic technologies with a supramolecular synthetic strategy, we present a high-throughput approach for formulating and screening multifunctional supramolecular nanoparticles (MFSNPs) self-assembled from a collection of functional modules to achieve simultaneous delivery of one gene and TF with unprecedented efficiency both in vitro and in vivo. We envision that this new approach could open a new avenue for immunotherapy, stem cell reprogramming, and other therapeutic applications.
Collapse
Affiliation(s)
- Yang Liu
- Laboratory of Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Chinese Academy of Sciences, No. 11 Beiyitiao, Zhongguancun, Beijing (China)
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin (China)
- California NanoSystems Institute, Department of Chemical and Bimolecular Engineering, UCLA (USA)
| | - Juanjuan Du
- California NanoSystems Institute, Department of Chemical and Bimolecular Engineering, UCLA (USA)
| | - Jin-sil Choi
- Crump Institute for Molecular Imaging, California NanoSystems Institute, Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA 90095 (USA)
| | - Kuan-Ju Chen
- Crump Institute for Molecular Imaging, California NanoSystems Institute, Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA 90095 (USA)
| | - Shuang Hou
- Crump Institute for Molecular Imaging, California NanoSystems Institute, Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA 90095 (USA)
| | - Ming Yan
- California NanoSystems Institute, Department of Chemical and Bimolecular Engineering, UCLA (USA)
| | - Wei-Yu Lin
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, 80708 (Taiwan)
| | - Kevin Sean Chen
- Crump Institute for Molecular Imaging, California NanoSystems Institute, Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA 90095 (USA)
| | - Tracy Ro
- Crump Institute for Molecular Imaging, California NanoSystems Institute, Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA 90095 (USA)
| | - Gerald S Lipshutz
- Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine, UCLA (USA)
| | - Lily Wu
- Crump Institute for Molecular Imaging, California NanoSystems Institute, Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA 90095 (USA)
| | - Linqi Shi
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin (China)
| | - Yunfeng Lu
- California NanoSystems Institute, Department of Chemical and Bimolecular Engineering, UCLA (USA).
| | - Hsian-Rong Tseng
- Crump Institute for Molecular Imaging, California NanoSystems Institute, Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA 90095 (USA).
| | - Hao Wang
- Laboratory of Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Chinese Academy of Sciences, No. 11 Beiyitiao, Zhongguancun, Beijing (China).
| |
Collapse
|
37
|
Liu Y, Du J, Choi JS, Chen KJ, Hou S, Yan M, Lin WY, Chen KS, Ro T, Lipshutz GS, Wu L, Shi L, Lu Y, Tseng HR, Wang H. A High-Throughput Platform for Formulating and Screening Multifunctional Nanoparticles Capable of Simultaneous Delivery of Genes and Transcription Factors. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201507546] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
38
|
Abstract
The underlying physical properties of microfluidic tools have led to new biological insights through the development of microsystems that can manipulate, mimic and measure biology at a resolution that has not been possible with macroscale tools. Microsystems readily handle sub-microlitre volumes, precisely route predictable laminar fluid flows and match both perturbations and measurements to the length scales and timescales of biological systems. The advent of fabrication techniques that do not require highly specialized engineering facilities is fuelling the broad dissemination of microfluidic systems and their adaptation to specific biological questions. We describe how our understanding of molecular and cell biology is being and will continue to be advanced by precision microfluidic approaches and posit that microfluidic tools - in conjunction with advanced imaging, bioinformatics and molecular biology approaches - will transform biology into a precision science.
Collapse
|
39
|
Furnari FB, Cloughesy TF, Cavenee WK, Mischel PS. Heterogeneity of epidermal growth factor receptor signalling networks in glioblastoma. Nat Rev Cancer 2015; 15:302-10. [PMID: 25855404 PMCID: PMC4875778 DOI: 10.1038/nrc3918] [Citation(s) in RCA: 286] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
As tumours evolve, the daughter cells of the initiating cell often become molecularly heterogeneous and develop different functional properties and therapeutic vulnerabilities. In glioblastoma (GBM), a lethal form of brain cancer, the heterogeneous expression of the epidermal growth factor receptor (EGFR) poses a substantial challenge for the effective use of EGFR-targeted therapies. Understanding the mechanisms that cause EGFR heterogeneity in GBM should provide better insights into how they, and possibly other amplified receptor tyrosine kinases, affect cellular signalling, metabolism and drug resistance.
Collapse
Affiliation(s)
- Frank B Furnari
- Ludwig Institute for Cancer Research and the Department of Pathology, University of California San Diego, La Jolla, California 92093, USA
| | - Timothy F Cloughesy
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, California 90095, USA
| | - Webster K Cavenee
- Ludwig Institute for Cancer Research and the Department of Medicine, University of California San Diego, La Jolla, California 92093, USA
| | - Paul S Mischel
- Ludwig Institute for Cancer Research and the Department of Pathology, University of California San Diego, La Jolla, California 92093, USA
| |
Collapse
|
40
|
Galler K, Bräutigam K, Große C, Popp J, Neugebauer U. Making a big thing of a small cell--recent advances in single cell analysis. Analyst 2015; 139:1237-73. [PMID: 24495980 DOI: 10.1039/c3an01939j] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Single cell analysis is an emerging field requiring a high level interdisciplinary collaboration to provide detailed insights into the complex organisation, function and heterogeneity of life. This review is addressed to life science researchers as well as researchers developing novel technologies. It covers all aspects of the characterisation of single cells (with a special focus on mammalian cells) from morphology to genetics and different omics-techniques to physiological, mechanical and electrical methods. In recent years, tremendous advances have been achieved in all fields of single cell analysis: (1) improved spatial and temporal resolution of imaging techniques to enable the tracking of single molecule dynamics within single cells; (2) increased throughput to reveal unexpected heterogeneity between different individual cells raising the question what characterizes a cell type and what is just natural biological variation; and (3) emerging multimodal approaches trying to bring together information from complementary techniques paving the way for a deeper understanding of the complexity of biological processes. This review also covers the first successful translations of single cell analysis methods to diagnostic applications in the field of tumour research (especially circulating tumour cells), regenerative medicine, drug discovery and immunology.
Collapse
Affiliation(s)
- Kerstin Galler
- Integrated Research and Treatment Center "Center for Sepsis Control and Care", Jena University Hospital, Erlanger Allee 101, 07747 Jena, Germany
| | | | | | | | | |
Collapse
|
41
|
Qiu X, De Jesus J, Pennell M, Troiani M, Haun JB. Microfluidic device for mechanical dissociation of cancer cell aggregates into single cells. LAB ON A CHIP 2015; 15:339-350. [PMID: 25377468 PMCID: PMC4301619 DOI: 10.1039/c4lc01126k] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Tumors tissues house a diverse array of cell types, requiring powerful cell-based analysis methods to characterize cellular heterogeneity and identify rare cells. Tumor tissue is dissociated into single cells by treatment with proteolytic enzymes, followed by mechanical disruption using vortexing or pipetting. These procedures can be incomplete and require significant time, and the latter mechanical treatments are poorly defined and controlled. Here, we present a novel microfluidic device to improve mechanical dissociation of digested tissue and cell aggregates into single cells. The device design includes a network of branching channels that range in size from millimeters down to hundreds of microns. The channels also contain flow constrictions that generate well-defined regions of high shear force, which we refer to as "hydrodynamic micro-scalpels", to progressively disaggregate tissue fragments and clusters into single cells. We show using in vitro cancer cell models that the microfluidic device significantly enhances cell recovery in comparison to mechanical disruption by pipetting and vortexing after digestion with trypsin or incubation with EDTA. Notably, the device enabled superior results to be obtained after shorter proteolytic digestion times, resulting in fully viable cells in less than ten minutes. The device could also be operated under enzyme-free conditions that could better maintain expression of certain surface markers. The microfluidic format is advantageous because it enables application of well-defined mechanical forces and rapid processing times. Furthermore, it may be possible to directly integrate downstream processing and detection operations to create integrated cell-based analysis platforms. The enhanced capabilities enabled by our novel device may help promote applications of single cell detection and purification techniques to tumor tissue specimens, advancing the current understanding of cancer biology and enabling molecular diagnostics in clinical settings.
Collapse
Affiliation(s)
- Xiaolong Qiu
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697
| | - Janice De Jesus
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697
| | - Marissa Pennell
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697
| | - Marco Troiani
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697
| | - Jered B. Haun
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697
- Department of Chemical Engineering and Materials Science, University of California Irvine, Irvine, CA 92697
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA 92697
- Corresponding Author: Jered B. Haun, PhD, Department of Biomedical Engineering University of California Irvine 3107 Natural Sciences II, Irvine, CA, 92697 949-824-1243,
| |
Collapse
|
42
|
Nahavandi S, Tang SY, Baratchi S, Soffe R, Nahavandi S, Kalantar-zadeh K, Mitchell A, Khoshmanesh K. Microfluidic platforms for the investigation of intercellular signalling mechanisms. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2014; 10:4810-26. [PMID: 25238429 DOI: 10.1002/smll.201401444] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 06/27/2014] [Indexed: 05/02/2023]
Abstract
Intercellular signalling has been identified as a highly complex process, responsible for orchestrating many physiological functions. While conventional methods of investigation have been useful, their limitations are impeding further development. Microfluidics offers an opportunity to overcome some of these limitations. Most notably, microfluidic systems can emulate the in-vivo environments. Further, they enable exceptionally precise control of the microenvironment, allowing complex mechanisms to be selectively isolated and studied in detail. There has thus been a growing adoption of microfluidic platforms for investigation of cell signalling mechanisms. This review provides an overview of the different signalling mechanisms and discusses the methods used to study them, with a focus on the microfluidic devices developed for this purpose.
Collapse
Affiliation(s)
- Sofia Nahavandi
- Faculty of Medicine, Dentistry, & Health Sciences, The University of Melbourne, VIC 3010, Australia
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Lin M, Chen JF, Lu YT, Zhang Y, Song J, Hou S, Ke Z, Tseng HR. Nanostructure embedded microchips for detection, isolation, and characterization of circulating tumor cells. Acc Chem Res 2014; 47:2941-50. [PMID: 25111636 PMCID: PMC4204926 DOI: 10.1021/ar5001617] [Citation(s) in RCA: 167] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Circulating
tumor cells (CTCs) are cancer cells that break away
from either a primary tumor or a metastatic site and circulate in
the peripheral blood as the cellular origin of metastasis. With their
role as a “tumor liquid biopsy”, CTCs provide convenient
access to all disease sites, including that of the primary tumor and
the site of fatal metastases. It is conceivable that detecting and
analyzing CTCs will provide insightful information in assessing the
disease status without the flaws and limitations encountered in performing
conventional tumor biopsies. However, identifying CTCs in patient
blood samples is technically challenging due to the extremely low
abundance of CTCs among a large number of hematologic cells. To address
this unmet need, there have been significant research endeavors, especially
in the fields of chemistry, materials science, and bioengineering,
devoted to developing CTC detection, isolation, and characterization
technologies. Inspired by the nanoscale interactions observed
in the tissue microenvironment,
our research team at UCLA pioneered a unique concept of “NanoVelcro”
cell-affinity substrates, in which CTC capture agent-coated nanostructured
substrates were utilized to immobilize CTCs with high efficiency.
The working mechanism of NanoVelcro cell-affinity substrates mimics
that of Velcro: when the two fabric strips of a Velcro fastener are
pressed together, tangling between the hairy surfaces on two strips
leads to strong binding. Through continuous evolution, three generations
(gens) of NanoVelcro CTC chips have been established to achieve different
clinical utilities. The first-gen NanoVelcro chip, composed of a silicon
nanowire substrate (SiNS) and an overlaid microfluidic chaotic mixer,
was created for CTC enumeration. Side-by-side analytical validation
studies using clinical blood samples suggested that the sensitivity
of first-gen NanoVelcro chip outperforms that of FDA-approved CellSearch.
In conjunction with the use of the laser microdissection (LMD) technique,
second-gen NanoVelcro chips (i.e., NanoVelcro-LMD), based on polymer
nanosubstrates, were developed for single-CTC isolation. The individually
isolated CTCs can be subjected to single-CTC genotyping (e.g., Sanger
sequencing and next-generation sequencing, NGS) to verify the CTC’s
role as tumor liquid biopsy. Created by grafting of thermoresponsive
polymer brushes onto SiNS, third-gen NanoVelcro chips (i.e., Thermoresponsive
NanoVelcro) have demonstrated the capture and release of CTCs at 37
and 4 °C, respectively. The temperature-dependent conformational
changes of polymer brushes can effectively alter the accessibility
of the capture agent on SiNS, allowing for rapid CTC purification
with desired viability and molecular integrity. This Account
summarizes the continuous evolution of NanoVelcro
CTC assays from the emergence of the original idea all the way to
their applications in cancer research. We envision that NanoVelcro
CTC assays will lead the way for powerful and cost-efficient diagnostic
platforms for researchers to better understand underlying disease
mechanisms and for physicians to monitor real-time disease progression.
Collapse
Affiliation(s)
- Millicent Lin
- Department
of Pathology, The First Affiliated hospital of Sun Yat-sen University, Guangzhou, 510080 Guangdong, People’s Republic of China
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| | - Jie-Fu Chen
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| | - Yi-Tsung Lu
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| | - Yang Zhang
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| | - Jinzhao Song
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| | - Shuang Hou
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| | - Zunfu Ke
- Department
of Pathology, The First Affiliated hospital of Sun Yat-sen University, Guangzhou, 510080 Guangdong, People’s Republic of China
| | - Hsian-Rong Tseng
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| |
Collapse
|
44
|
Glioblastoma multiforme: a look inside its heterogeneous nature. Cancers (Basel) 2014; 6:226-39. [PMID: 24473088 PMCID: PMC3980595 DOI: 10.3390/cancers6010226] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/23/2013] [Accepted: 01/09/2014] [Indexed: 11/17/2022] Open
Abstract
Heterogeneity is a hallmark of tumors and has a crucial role in the outcome of the malignancy, because it not only confounds diagnosis, but also challenges the design of effective therapies. There are two types of heterogeneity: inter-tumor and intra-tumor heterogeneity. While inter-tumor heterogeneity has been studied widely, intra-tumor heterogeneity has been neglected even though numerous studies support this aspect of tumor pathobiology. The main reason has been the technical difficulties, but with new advances in single-cell technology, intra-tumor heterogeneity is becoming a key area in the study of cancer. Several models try to explain the origin and maintenance of intra-tumor heterogeneity, however, one prominent model compares cancer with a tree where the ubiquitous mutations compose the trunk and mutations present in subpopulations of cells are represented by the branches. In this review we will focus on the intra-tumor heterogeneity of glioblastoma multiforme (GBM), the most common brain tumor in adults that is characterized by a marked heterogeneity at the cellular and molecular levels. Better understanding of this heterogeneity will be essential to design effective therapies against this devastating disease to avoid tumor escape.
Collapse
|
45
|
Yu J, Zhou J, Sutherland A, Wei W, Shin YS, Xue M, Heath JR. Microfluidics-based single-cell functional proteomics for fundamental and applied biomedical applications. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2014; 7:275-95. [PMID: 24896308 DOI: 10.1146/annurev-anchem-071213-020323] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
We review an emerging microfluidics-based toolkit for single-cell functional proteomics. Functional proteins include, but are not limited to, the secreted signaling proteins that can reflect the biological behaviors of immune cells or the intracellular phosphoproteins associated with growth factor-stimulated signaling networks. Advantages of the microfluidics platforms are multiple. First, 20 or more functional proteins may be assayed simultaneously from statistical numbers of single cells. Second, cell behaviors (e.g., motility) may be correlated with protein assays. Third, extensions to quantized cell populations can permit measurements of cell-cell interactions. Fourth, rare cells can be functionally identified and then separated for further analysis or culturing. Finally, certain assay types can provide a conduit between biology and the physicochemical laws. We discuss the history and challenges of the field then review design concepts and uses of the microchip platforms that have been reported, with an eye toward biomedical applications. We then look to the future of the field.
Collapse
Affiliation(s)
- Jing Yu
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125;
| | | | | | | | | | | | | |
Collapse
|
46
|
Nathanson DA, Gini B, Mottahedeh J, Visnyei K, Koga T, Gomez G, Eskin A, Hwang K, Wang J, Masui K, Paucar A, Yang H, Ohashi M, Zhu S, Wykosky J, Reed R, Nelson SF, Cloughesy TF, James CD, Rao PN, Kornblum HI, Heath JR, Cavenee WK, Furnari FB, Mischel PS. Targeted therapy resistance mediated by dynamic regulation of extrachromosomal mutant EGFR DNA. Science 2013; 343:72-6. [PMID: 24310612 DOI: 10.1126/science.1241328] [Citation(s) in RCA: 423] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Intratumoral heterogeneity contributes to cancer drug resistance, but the underlying mechanisms are not understood. Single-cell analyses of patient-derived models and clinical samples from glioblastoma patients treated with epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) demonstrate that tumor cells reversibly up-regulate or suppress mutant EGFR expression, conferring distinct cellular phenotypes to reach an optimal equilibrium for growth. Resistance to EGFR TKIs is shown to occur by elimination of mutant EGFR from extrachromosomal DNA. After drug withdrawal, reemergence of clonal EGFR mutations on extrachromosomal DNA follows. These results indicate a highly specific, dynamic, and adaptive route by which cancers can evade therapies that target oncogenes maintained on extrachromosomal DNA.
Collapse
Affiliation(s)
- David A Nathanson
- Ludwig Institute for Cancer Research, University of California at San Diego, La Jolla, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lu YT, Zhao L, Shen Q, Garcia MA, Wu D, Hou S, Song M, Xu X, OuYang WH, OuYang WWL, Lichterman J, Luo Z, Xuan X, Huang J, Chung LWK, Rettig M, Tseng HR, Shao C, Posadas EM. NanoVelcro Chip for CTC enumeration in prostate cancer patients. Methods 2013; 64:144-52. [PMID: 23816790 PMCID: PMC3834112 DOI: 10.1016/j.ymeth.2013.06.019] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 05/30/2013] [Accepted: 06/18/2013] [Indexed: 01/18/2023] Open
Abstract
Circulating tumor cells (CTCs) are one of the most crucial topics in rare cell biology and have become the focus of a significant and emerging area of cancer research. While CTC enumeration is a valid biomarker in prostate cancer, the current FDA-approved CTC technology is unable to detect CTCs in a large portion of late stage prostate cancer patients. Here we introduce the NanoVelcro CTC Chip, a device composed of a patterned silicon nanowire substrate (SiNW) and an overlaid polydimethylsiloxane (PDMS) chaotic mixer. Validated by two institutions participating in the study, the NanoVelcro Chip assay exhibits very consistent efficiency in CTC-capture from patient samples. The utilized protocol can be easily replicated at different facilities. We demonstrate the clinical utility of the NanoVelcro Chip by performing serial enumerations of CTCs in prostate cancer patients after undergoing systemic therapy. Changes in CTC numbers after 4-10 weeks of therapy were compared with their clinical responses. We observed a statistically significant reduction in CTCs counts in the clinical responders. We performed long-term follow up with serial CTC collection and enumeration in one patient observing variations in counts correlating with treatment response. This study demonstrates the consistency of the NanoVelcro Chip assay over time for CTC enumeration and also shows that continuous monitoring of CTC numbers can be employed to follow responses to different treatments and monitor disease progression.
Collapse
Affiliation(s)
- Yi-Tsung Lu
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Urologic Oncology Program & Uro-Oncology Research Program, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Libo Zhao
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
- Crump Institute for Molecular Imaging (CIMI), University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA 90095, USA
| | - Qinglin Shen
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
- Crump Institute for Molecular Imaging (CIMI), University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA 90095, USA
| | - Mitch A. Garcia
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
- Crump Institute for Molecular Imaging (CIMI), University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA 90095, USA
| | - Dongxia Wu
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
- Crump Institute for Molecular Imaging (CIMI), University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA 90095, USA
| | - Shuang Hou
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
- Crump Institute for Molecular Imaging (CIMI), University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA 90095, USA
| | - Min Song
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
- Crump Institute for Molecular Imaging (CIMI), University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA 90095, USA
| | - Xiaochun Xu
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
- Crump Institute for Molecular Imaging (CIMI), University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA 90095, USA
| | - Wei-Han OuYang
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
- Crump Institute for Molecular Imaging (CIMI), University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA 90095, USA
| | - William W.-L. OuYang
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
- Crump Institute for Molecular Imaging (CIMI), University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA 90095, USA
| | - Jake Lichterman
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Urologic Oncology Program & Uro-Oncology Research Program, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Zheng Luo
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
- Crump Institute for Molecular Imaging (CIMI), University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA 90095, USA
| | - Xuan Xuan
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
- Crump Institute for Molecular Imaging (CIMI), University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA 90095, USA
| | - Jiaoti Huang
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095, USA
| | - Leland W. K. Chung
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Urologic Oncology Program & Uro-Oncology Research Program, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Matthew Rettig
- Division of Hematology/Oncology, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- Departments of Medicine and Urology, Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Hsian-Rong Tseng
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
- Crump Institute for Molecular Imaging (CIMI), University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, CA 90095, USA
| | - Chen Shao
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Urologic Oncology Program & Uro-Oncology Research Program, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, P. R. China
| | - Edwin M. Posadas
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Urologic Oncology Program & Uro-Oncology Research Program, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
48
|
Qureshi IA, Mehler MF. Developing epigenetic diagnostics and therapeutics for brain disorders. Trends Mol Med 2013; 19:732-41. [PMID: 24145019 DOI: 10.1016/j.molmed.2013.09.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 08/30/2013] [Accepted: 09/19/2013] [Indexed: 12/11/2022]
Abstract
Perturbations in epigenetic mechanisms have emerged as cardinal features in the molecular pathology of major classes of brain disorders. We therefore highlight evidence which suggests that specific epigenetic signatures measurable in central - and possibly even in peripheral tissues - have significant value as translatable biomarkers for screening, early diagnosis, and prognostication; developing molecularly targeted medicines; and monitoring disease progression and treatment responses. We also draw attention to existing and novel therapeutic approaches directed at epigenetic factors and mechanisms, including strategies for modulating enzymes that write and erase DNA methylation and histone/chromatin marks; protein-protein interactions responsible for reading epigenetic marks; and non-coding RNA pathways.
Collapse
Affiliation(s)
- Irfan A Qureshi
- Roslyn and Leslie Goldstein Laboratory for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Rose F. Kennedy Center for Research on Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | |
Collapse
|
49
|
Microfluidic chip-based technologies: emerging platforms for cancer diagnosis. BMC Biotechnol 2013; 13:76. [PMID: 24070124 PMCID: PMC3849190 DOI: 10.1186/1472-6750-13-76] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Accepted: 09/05/2013] [Indexed: 12/20/2022] Open
Abstract
The development of early and personalized diagnostic protocols is considered the most promising avenue to decrease mortality from cancer and improve outcome. The emerging microfluidic-based analyzing platforms hold high promises to fulfill high-throughput and high-precision screening with reduced equipment cost and low analysis time, as compared to traditional bulky counterparts in bench-top laboratories. This article overviewed the potential applications of microfluidic technologies for detection and monitoring of cancer through nucleic acid and protein biomarker analysis. The implications of the technologies in cancer cytology that can provide functional personalized diagnosis were highlighted. Finally, the future niches for using microfluidic-based systems in tumor screening were briefly discussed.
Collapse
|
50
|
Ly J, Masterman-Smith M, Ramakrishnan R, Sun J, Kokubun B, van Dam RM. Automated reagent-dispensing system for microfluidic cell biology assays. ACTA ACUST UNITED AC 2013; 18:530-41. [PMID: 24051515 DOI: 10.1177/2211068213504758] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Microscale systems that enable measurements of oncological phenomena at the single-cell level have a great capacity to improve therapeutic strategies and diagnostics. Such measurements can reveal unprecedented insights into cellular heterogeneity and its implications into the progression and treatment of complicated cellular disease processes such as those found in cancer. We describe a novel fluid-delivery platform to interface with low-cost microfluidic chips containing arrays of microchambers. Using multiple pairs of needles to aspirate and dispense reagents, the platform enables automated coating of chambers, loading of cells, and treatment with growth media or other agents (e.g., drugs, fixatives, membrane permeabilizers, washes, stains, etc.). The chips can be quantitatively assayed using standard fluorescence-based immunocytochemistry, microscopy, and image analysis tools, to determine, for example, drug response based on differences in protein expression and/or activation of cellular targets on an individual-cell level. In general, automation of fluid and cell handling increases repeatability, eliminates human error, and enables increased throughput, especially for sophisticated, multistep assays such as multiparameter quantitative immunocytochemistry. We report the design of the automated platform and compare several aspects of its performance to manually-loaded microfluidic chips.
Collapse
Affiliation(s)
- Jimmy Ly
- 1Department of Bioengineering, Samueli School of Engineering and Applied Sciences, University of California, Los Angeles, CA, USA
| | | | | | | | | | | |
Collapse
|