1
|
Shi Y, Han X, Zou S, Liu G. Nanomaterials in Organoids: From Interactions to Personalized Medicine. ACS NANO 2024; 18:33276-33292. [PMID: 39609736 DOI: 10.1021/acsnano.4c13330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Organoids are three-dimensional models of microscopic organisms created through the self-organization of various types of stem cells. They are widely unitized in personalized medicine due to their capacity to replicate the structure and functionality of native organs. Meanwhile, nanotechnology has been integrated into diagnostic and therapeutic tools to manage an array of medical conditions, given its unique characteristics of nanoscale. Nanomaterials have demonstrated potential in developing innovative and effective organoids. With a focus on studying the interaction of nanomaterials and organoid technology in personalized medicine, this Review examines the role of nanomaterials in regulating the fate of stem cells to construct different types of organoids. It also explores the potential of nanotechnology to create 3D microenvironments for organoids. Finally, perspectives and challenges of applying nanotechnology for organoids development toward the translation of personalized medicine are discussed.
Collapse
Affiliation(s)
- Ying Shi
- Integrated Devices and Intelligent Diagnosis (ID2) Laboratory, CUHK(SZ)-Boyalife Joint Laboratory of Regenerative Medicine Engineering, Biomedical Engineering Programme, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Xin Han
- Integrated Devices and Intelligent Diagnosis (ID2) Laboratory, CUHK(SZ)-Boyalife Joint Laboratory of Regenerative Medicine Engineering, Biomedical Engineering Programme, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Siyi Zou
- Integrated Devices and Intelligent Diagnosis (ID2) Laboratory, CUHK(SZ)-Boyalife Joint Laboratory of Regenerative Medicine Engineering, Biomedical Engineering Programme, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Guozhen Liu
- Integrated Devices and Intelligent Diagnosis (ID2) Laboratory, CUHK(SZ)-Boyalife Joint Laboratory of Regenerative Medicine Engineering, Biomedical Engineering Programme, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| |
Collapse
|
2
|
Azuaje-Hualde E, Alonso-Cabrera JA, de Pancorbo MM, Benito-Lopez F, Basabe-Desmonts L. Integration of secreted signaling molecule sensing on cell monitoring platforms: a critical review. Anal Bioanal Chem 2024; 416:7249-7266. [PMID: 39048740 PMCID: PMC11584473 DOI: 10.1007/s00216-024-05435-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/10/2024] [Accepted: 06/28/2024] [Indexed: 07/27/2024]
Abstract
Monitoring cell secretion in complex microenvironments is crucial for understanding cellular behavior and advancing physiological and pathological research. While traditional cell culture methods, including organoids and spheroids, provide valuable models, real-time monitoring of cell secretion of signaling molecules remains challenging. Integrating advanced monitoring technologies into these systems often disrupts the delicate balance of the microenvironment, making it difficult to achieve sensitivity and specificity. This review explored recent strategies for integrating the monitoring of cell secretion of signaling molecules, crucial for understanding and replicating cell microenvironments, within cell culture platforms, addressing challenges such as non-adherent cell models and the focus on single-cell methodologies. We highlight advancements in biosensors, microfluidics, and three-dimensional culture methods, and discuss their potential to enhance real-time, multiplexed cell monitoring. By examining the advantages, limitations, and future prospects of these technologies, we aim to contribute to the development of integrated systems that facilitate comprehensive cell monitoring, ultimately advancing biological research and pharmaceutical development.
Collapse
Affiliation(s)
- Enrique Azuaje-Hualde
- Microfluidics Cluster UPV/EHU, BIOMICs Microfluidics Group, Lascaray Research Center, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Juncal A Alonso-Cabrera
- Microfluidics Cluster UPV/EHU, BIOMICs Microfluidics Group, Lascaray Research Center, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Marian M de Pancorbo
- BIOMICs Research Group, Lascaray Research Center, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Fernando Benito-Lopez
- Microfluidics Cluster UPV/EHU, Analytical Microsystems & Materials for Lab-on-a-Chip (AMMa-LOAC) Group, Analytical Chemistry Department, University of the Basque Country UPV/EHU, Leioa, Spain.
- Microfluidics Cluster UPV/EHU, Bioaraba Health Research Institute, Vitoria-Gasteiz, Spain.
- Basque Foundation of Science, IKERBASQUE, María Díaz Haroko Kalea, 3, 48013, Bilbao, Spain.
| | - Lourdes Basabe-Desmonts
- Microfluidics Cluster UPV/EHU, BIOMICs Microfluidics Group, Lascaray Research Center, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain.
- Microfluidics Cluster UPV/EHU, Bioaraba Health Research Institute, Vitoria-Gasteiz, Spain.
- Basque Foundation of Science, IKERBASQUE, María Díaz Haroko Kalea, 3, 48013, Bilbao, Spain.
| |
Collapse
|
3
|
Zhang X, Su R, Wang H, Wu R, Fan Y, Bin Z, Gao C, Wang C. The promise of Synovial Joint-on-a-Chip in rheumatoid arthritis. Front Immunol 2024; 15:1408501. [PMID: 39324139 PMCID: PMC11422143 DOI: 10.3389/fimmu.2024.1408501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
Rheumatoid arthritis (RA) affects millions of people worldwide, but there are limited drugs available to treat it, so acquiring a more comprehensive comprehension of the underlying reasons and mechanisms behind inflammation is crucial, as well as developing novel therapeutic approaches to manage it and mitigate or forestall associated harm. It is evident that current in vitro models cannot faithfully replicate all aspects of joint diseases, which makes them ineffective as tools for disease research and drug testing. Organ-on-a-chip (OoC) technology is an innovative platform that can mimic the microenvironment and physiological state of living tissues more realistically than traditional methods by simulating the spatial arrangement of cells and interorgan communication. This technology allows for the precise control of fluid flow, nutrient exchange, and the transmission of physicochemical signals, such as bioelectrical, mechanical stimulation and shear force. In addition, the integration of cutting-edge technologies like sensors, 3D printing, and artificial intelligence enhances the capabilities of these models. Here, we delve into OoC models with a particular focus on Synovial Joints-on-a-Chip, where we outline their structure and function, highlighting the potential of the model to advance our understanding of RA. We integrate the actual evidence regarding various OoC models and their possible integration for multisystem disease study in RA research for the first time and introduce the prospects and opportunities of the chip in RA etiology and pathological mechanism research, drug research, disease prevention and human precision medicine. Although many challenges remain, OoC holds great promise as an in vitro model that approaches physiology and dynamics.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory for Immunomicroecology, Taiyuan, Shanxi, China
- Shanxi Province Engineering Research Center of Precision Medicine for Rheumatology, Taiyuan, Shanxi, China
| | - Rui Su
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory for Immunomicroecology, Taiyuan, Shanxi, China
- Shanxi Province Engineering Research Center of Precision Medicine for Rheumatology, Taiyuan, Shanxi, China
| | - Hui Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory for Immunomicroecology, Taiyuan, Shanxi, China
- Shanxi Province Engineering Research Center of Precision Medicine for Rheumatology, Taiyuan, Shanxi, China
| | - Ruihe Wu
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory for Immunomicroecology, Taiyuan, Shanxi, China
- Shanxi Province Engineering Research Center of Precision Medicine for Rheumatology, Taiyuan, Shanxi, China
| | - Yuxin Fan
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory for Immunomicroecology, Taiyuan, Shanxi, China
- Shanxi Province Engineering Research Center of Precision Medicine for Rheumatology, Taiyuan, Shanxi, China
| | - Zexuan Bin
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory for Immunomicroecology, Taiyuan, Shanxi, China
- Shanxi Province Engineering Research Center of Precision Medicine for Rheumatology, Taiyuan, Shanxi, China
| | - Chong Gao
- Pathology, Joint Program in Transfusion Medicine, Brigham and Women’s Hospital/Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Caihong Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory for Immunomicroecology, Taiyuan, Shanxi, China
- Shanxi Province Engineering Research Center of Precision Medicine for Rheumatology, Taiyuan, Shanxi, China
| |
Collapse
|
4
|
Spitz S, Schobesberger S, Brandauer K, Ertl P. Sensor-integrated brain-on-a-chip platforms: Improving the predictive validity in neurodegenerative research. Bioeng Transl Med 2024; 9:e10604. [PMID: 38818126 PMCID: PMC11135156 DOI: 10.1002/btm2.10604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/31/2023] [Accepted: 09/12/2023] [Indexed: 06/01/2024] Open
Abstract
Affecting millions of individuals worldwide, neurodegenerative diseases (NDDs) pose a significant and growing health concern in people over the age of 60 years. Contributing to this trend are the steady increase in the aging population coupled with a persistent lack of disease-altering treatment strategies targeting NDDs. The absence of efficient therapeutics can be attributed to high failure rates in clinical trials and the ineptness of animal models in preceding preclinical studies. To that end, in recent years, significant research effort has been dedicated to the development of human cell-based preclinical disease models characterized by a higher degree of predictive validity. However, a key requirement of any in vitro model constitutes the precise knowledge and replication of the target tissues' (patho-)physiological microenvironment. Herein, microphysiological systems have demonstrated superiority over conventional static 2D/3D in vitro cell culture systems, as they allow for the emulation and continuous monitoring of the onset, progression, and remission of disease-associated phenotypes. This review provides an overview of recent advances in the field of NDD research using organ-on-a-chip platforms. Specific focus is directed toward non-invasive sensing strategies encompassing electrical, electrochemical, and optical sensors. Additionally, promising on- and integrable off-chip sensing strategies targeting key analytes in NDDs will be presented and discussed in detail.
Collapse
Affiliation(s)
- Sarah Spitz
- Faculty of Technical ChemistryVienna University of TechnologyViennaAustria
- Present address:
Department of Mechanical Engineering and Biological EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | | | | | - Peter Ertl
- Faculty of Technical ChemistryVienna University of TechnologyViennaAustria
| |
Collapse
|
5
|
Tawade P, Mastrangeli M. Integrated Electrochemical and Optical Biosensing in Organs-on-Chip. Chembiochem 2024; 25:e202300560. [PMID: 37966365 DOI: 10.1002/cbic.202300560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/13/2023] [Accepted: 11/13/2023] [Indexed: 11/16/2023]
Abstract
Demand for biocompatible, non-invasive, and continuous real-time monitoring of organs-on-chip has driven the development of a variety of novel sensors. However, highest accuracy and sensitivity can arguably be achieved by integrated biosensing, which enables in situ monitoring of the in vitro microenvironment and dynamic responses of tissues and miniature organs recapitulated in organs-on-chip. This paper reviews integrated electrical, electrochemical, and optical sensing methods within organ-on-chip devices and platforms. By affording precise detection of analytes and biochemical reactions, these methods expand and advance the monitoring capabilities and reproducibility of organ-on-chip technology. The integration of these sensing techniques allows a deeper understanding of organ functions, and paves the way for important applications such as drug testing, disease modeling, and personalized medicine. By consolidating recent advancements and highlighting challenges in the field, this review aims to foster further research and innovation in the integration of biosensing in organs-on-chip.
Collapse
Affiliation(s)
- Pratik Tawade
- Electronic Components, Technology and Materials, Department of Microelectronics, Delft University of Technology, Mekelweg 4, 2628CD, Delft, Netherlands
| | - Massimo Mastrangeli
- Electronic Components, Technology and Materials, Department of Microelectronics, Delft University of Technology, Mekelweg 4, 2628CD, Delft, Netherlands
| |
Collapse
|
6
|
Aruwa CE, Sabiu S. Adipose tissue inflammation linked to obesity: A review of current understanding, therapies and relevance of phyto-therapeutics. Heliyon 2024; 10:e23114. [PMID: 38163110 PMCID: PMC10755291 DOI: 10.1016/j.heliyon.2023.e23114] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/25/2023] [Accepted: 11/27/2023] [Indexed: 01/03/2024] Open
Abstract
Obesity is a current global challenge affecting all ages and is characterized by the up-regulated secretion of bioactive factors/pathways which result in adipose tissue inflammation (ATI). Current obesity therapies are mainly focused on lifestyle (diet/nutrition) changes. This is because many chemosynthetic anti-obesogenic medications cause adverse effects like diarrhoea, dyspepsia, and faecal incontinence, among others. As such, it is necessary to appraise the efficacies and mechanisms of action of safer, natural alternatives like plant-sourced compounds, extracts [extractable phenol (EP) and macromolecular antioxidant (MA) extracts], and anti-inflammatory peptides, among others, with a view to providing a unique approach to obesity care. These natural alternatives may constitute potent therapies for ATI linked to obesity. The potential of MA compounds (analysed for the first time in this review) and extracts in ATI and obesity management is elucidated upon, while also highlighting research gaps and future prospects. Furthermore, immune cells, signalling pathways, genes, and adipocyte cytokines play key roles in ATI responses and are targeted in certain therapies. As a result, this review gives an in-depth appraisal of ATI linked to obesity, its causes, mechanisms, and effects of past, present, and future therapies for reversal and alleviation of ATI. Achieving a significant decrease in morbidity and mortality rates attributed to ATI linked to obesity and related comorbidities is possible as research improves our understanding over time.
Collapse
Affiliation(s)
- Christiana Eleojo Aruwa
- Department of Biotechnology and Food Science, Durban University of Technology, PO Box 1334, Durban, 4000, South Africa
| | - Saheed Sabiu
- Department of Biotechnology and Food Science, Durban University of Technology, PO Box 1334, Durban, 4000, South Africa
| |
Collapse
|
7
|
Medina A, Bruno J, Alemán JO. Metabolic flux analysis in adipose tissue reprogramming. IMMUNOMETABOLISM (COBHAM, SURREY) 2024; 6:e00039. [PMID: 38455681 PMCID: PMC10916752 DOI: 10.1097/in9.0000000000000039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 01/29/2024] [Indexed: 03/09/2024]
Abstract
Obesity is a growing epidemic in the United States and worldwide and is associated with insulin resistance and cardiovascular disease, among other comorbidities. Understanding of the pathology that links overnutrition to these disease processes is ongoing. Adipose tissue is a heterogeneous organ comprised of multiple different cell types and it is likely that dysregulated metabolism within these cell populations disrupts both inter- and intracellular interactions and is a key driver of human disease. In recent years, metabolic flux analysis, which offers a precise quantification of metabolic pathway fluxes in biological systems, has emerged as a candidate strategy for uncovering the metabolic changes that stoke these disease processes. In this mini review, we discuss metabolic flux analysis as an experimental tool, with a specific emphasis on mass spectrometry with isotope tracing as this is the technique most frequently used for metabolic flux analysis in adipocytes. Furthermore, we examine existing literature that uses metabolic flux analysis to further our understanding of adipose tissue biology. Our group has a specific interest in understanding the role of white adipose tissue inflammation in the progression of cardiometabolic disease, as we know that in obesity the accumulation of pro-inflammatory adipose tissue macrophages is associated with significant morbidity, so we use this as a paradigm throughout our review for framing the application of these experimental techniques. However, there are many other biological applications to which they can be applied to further understanding of not only adipose tissue biology but also systemic homeostasis.
Collapse
Affiliation(s)
- Ashley Medina
- Laboratory of Translational Obesity Research, New York University Grossman School of Medicine, New York, NY, USA
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Joanne Bruno
- Laboratory of Translational Obesity Research, New York University Grossman School of Medicine, New York, NY, USA
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - José O. Alemán
- Laboratory of Translational Obesity Research, New York University Grossman School of Medicine, New York, NY, USA
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
8
|
Gupta B, Malviya R, Srivastava S, Ahmad I, Rab SO, Uniyal P. Construction, Features and Regulatory Aspects of Organ-chip for Drug Delivery Applications: Advances and Prospective. Curr Pharm Des 2024; 30:1952-1965. [PMID: 38859792 DOI: 10.2174/0113816128305296240523112043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/25/2024] [Indexed: 06/12/2024]
Abstract
Organ-on-chip is an innovative technique that emerged from tissue engineering and microfluidic technologies. Organ-on-chip devices (OoCs) are anticipated to provide efficient explanations for dealing with challenges in pharmaceutical advancement and individualized illness therapies. Organ-on-chip is an advanced method that can replicate human organs' physiological conditions and functions on a small chip. It possesses the capacity to greatly transform the drug development process by enabling the simulation of diseases and the testing of drugs. Effective integration of this advanced technical platform with common pharmaceutical and medical contexts is still a challenge. Microfluidic technology, a micro-level technique, has become a potent tool for biomedical engineering research. As a result, it has revolutionized disciplines, including physiological material interpreting, compound detection, cell-based assay, tissue engineering, biological diagnostics, and pharmaceutical identification. This article aims to offer an overview of newly developed organ-on-a-chip systems. It includes single-organ platforms, emphasizing the most researched organs, including the heart, liver, blood arteries, and lungs. Subsequently, it provides a concise overview of tumor-on-a-chip systems and emphasizes their use in evaluating anti-cancer medications.
Collapse
Affiliation(s)
- Babita Gupta
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, U.P., India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, U.P., India
| | - Saurabh Srivastava
- School of Pharmacy, KPJ Healthcare University College (KPJUC), Nilai, Malaysia
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Prerna Uniyal
- School of Pharmacy, Graphic Era Hill University, Dehradun, India
| |
Collapse
|
9
|
He J, Wu S, Chen W, Kim A, Yang W, Wang C, Gu Z, Shen J, Dai S, Chen W, Chen P. Calligraphy of Nanoplasmonic Bioink-Based Multiplex Immunosensor for Precision Immune Monitoring and Modulation. ACS APPLIED MATERIALS & INTERFACES 2023; 15:50047-50057. [PMID: 37856877 PMCID: PMC11694655 DOI: 10.1021/acsami.3c11417] [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] [Indexed: 10/21/2023]
Abstract
Immunomodulation therapies have attracted immense interest recently for the treatment of immune-related diseases, such as cancer and viral infections. This new wave of enthusiasm for immunomodulators, predominantly revolving around cytokines, has spurred emerging needs and opportunities for novel immune monitoring and diagnostic tools. Considering the highly dynamic immune status and limited window for therapeutic intervention, precise real-time detection of cytokines is critical to effectively monitor and manage the immune system and optimize the therapeutic outcome. The clinical success of such a rapid, sensitive, multiplex immunoanalytical platform further requires the system to have ease of integration and fabrication for sample sparing and large-scale production toward massive parallel analysis. In this article, we developed a nanoplasmonic bioink-based, label-free, multiplex immunosensor that can be readily "written" onto a glass substrate via one-step calligraphy patterning. This facile nanolithography technique allows programmable patterning of a minimum of 3 μL of nanoplasmonic bioink in 1 min and thus enables fabrication of a nanoplasmonic microarray immunosensor with 2 h simple incubation. The developed immunosensor was successfully applied for real-time, parallel detection of multiple cytokines (e.g., interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), and transforming growth factor-beta (TGF-β)) in immunomodulated macrophage samples. This integrated platform synergistically incorporates the concepts of nanosynthesis, nanofabrication, and nanobiosensing, showing great potential in the scalable production of label-free multiplex immunosensing devices with superior analytical performance for clinical applications in immunodiagnostics and immunotherapy.
Collapse
Affiliation(s)
- Jiacheng He
- Materials Research and Education Center, Auburn University, Auburn, Alabama 36849, United States
| | - Siqi Wu
- Materials Research and Education Center, Auburn University, Auburn, Alabama 36849, United States
| | - Wu Chen
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, Alabama 36849, United States
| | - Albert Kim
- Materials Research and Education Center, Auburn University, Auburn, Alabama 36849, United States
- Center for Medicine, Health, and Society, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Wen Yang
- Materials Research and Education Center, Auburn University, Auburn, Alabama 36849, United States
| | - Chuanyu Wang
- Materials Research and Education Center, Auburn University, Auburn, Alabama 36849, United States
| | - Zhengyang Gu
- Materials Research and Education Center, Auburn University, Auburn, Alabama 36849, United States
| | - Jialiang Shen
- Materials Research and Education Center, Auburn University, Auburn, Alabama 36849, United States
| | - Siyuan Dai
- Materials Research and Education Center, Auburn University, Auburn, Alabama 36849, United States
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, New York University, New York, New York 11201, United States
- Department of Biomedical Engineering, New York University, Brooklyn, New York 11201, United States
| | - Pengyu Chen
- Materials Research and Education Center, Auburn University, Auburn, Alabama 36849, United States
| |
Collapse
|
10
|
Tabatabaei Rezaei N, Kumar H, Liu H, Lee SS, Park SS, Kim K. Recent Advances in Organ-on-Chips Integrated with Bioprinting Technologies for Drug Screening. Adv Healthc Mater 2023; 12:e2203172. [PMID: 36971091 PMCID: PMC11469032 DOI: 10.1002/adhm.202203172] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/27/2023] [Indexed: 03/29/2023]
Abstract
Currently, the demand for more reliable drug screening devices has made scientists and researchers develop novel potential approaches to offer an alternative to animal studies. Organ-on-chips are newly emerged platforms for drug screening and disease metabolism investigation. These microfluidic devices attempt to recapitulate the physiological and biological properties of different organs and tissues using human-derived cells. Recently, the synergistic combination of additive manufacturing and microfluidics has shown a promising impact on improving a wide array of biological models. In this review, different methods are classified using bioprinting to achieve the relevant biomimetic models in organ-on-chips, boosting the efficiency of these devices to produce more reliable data for drug investigations. In addition to the tissue models, the influence of additive manufacturing on microfluidic chip fabrication is discussed, and their biomedical applications are reviewed.
Collapse
Affiliation(s)
- Nima Tabatabaei Rezaei
- Department of Mechanical and Manufacturing EngineeringUniversity of CalgaryCalgaryAlbertaT2N 1N4Canada
| | - Hitendra Kumar
- Department of Mechanical and Manufacturing EngineeringUniversity of CalgaryCalgaryAlbertaT2N 1N4Canada
- Department of Pathology and Laboratory MedicineCumming School of MedicineUniversity of CalgaryCalgaryAlbertaT2N 1N4Canada
| | - Hongqun Liu
- Liver UnitCumming School of MedicineUniversity of CalgaryCalgaryAlbertaT2N 1N4Canada
| | - Samuel S. Lee
- Liver UnitCumming School of MedicineUniversity of CalgaryCalgaryAlbertaT2N 1N4Canada
| | - Simon S. Park
- Department of Mechanical and Manufacturing EngineeringUniversity of CalgaryCalgaryAlbertaT2N 1N4Canada
| | - Keekyoung Kim
- Department of Mechanical and Manufacturing EngineeringUniversity of CalgaryCalgaryAlbertaT2N 1N4Canada
- Department of Biomedical EngineeringUniversity of CalgaryCalgaryAlbertaT2N 1N4Canada
| |
Collapse
|
11
|
Zhang W, Zi X, Bi J, Liu G, Cheng H, Bao K, Qin L, Wang W. Plasmonic Nanomaterials in Dark Field Sensing Systems. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2027. [PMID: 37446543 DOI: 10.3390/nano13132027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/26/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023]
Abstract
Plasma nanoparticles offer promise in data storage, biosensing, optical imaging, photoelectric integration, etc. This review highlights the local surface plasmon resonance (LSPR) excitation mechanism of plasmonic nanoprobes and its critical significance in the control of dark-field sensing, as well as three main sensing strategies based on plasmonic nanomaterial dielectric environment modification, electromagnetic coupling, and charge transfer. This review then describes the component materials of plasmonic nanoprobes based on gold, silver, and other noble metals, as well as their applications. According to this summary, researchers raised the LSPR performance of composite plasmonic nanomaterials by combining noble metals with other metals or oxides and using them in process analysis and quantitative detection.
Collapse
Affiliation(s)
- Wenjia Zhang
- Tianjin Research Institute of Water Transport Engineering, M.O.T., Tianjin 300456, China
- National Engineering Research Center of Port Hydraulic Construction Technology, Tianjin 300456, China
| | - Xingyu Zi
- College of Microelectronics, Nankai University, Tianjin 300350, China
- Tianjin Key Laboratory of Optoelectronic Sensor and Sensing Network Technology, Tianjin 300350, China
| | - Jinqiang Bi
- Tianjin Research Institute of Water Transport Engineering, M.O.T., Tianjin 300456, China
- National Engineering Research Center of Port Hydraulic Construction Technology, Tianjin 300456, China
- School of Marine Science and Technology, Tianjin University, Tianjin 300192, China
| | - Guohua Liu
- College of Microelectronics, Nankai University, Tianjin 300350, China
- Tianjin Key Laboratory of Optoelectronic Sensor and Sensing Network Technology, Tianjin 300350, China
| | - Hongen Cheng
- College of Microelectronics, Nankai University, Tianjin 300350, China
- Tianjin Key Laboratory of Optoelectronic Sensor and Sensing Network Technology, Tianjin 300350, China
| | - Kexin Bao
- Tianjin Research Institute of Water Transport Engineering, M.O.T., Tianjin 300456, China
- National Engineering Research Center of Port Hydraulic Construction Technology, Tianjin 300456, China
- School of Marine Science and Technology, Tianjin University, Tianjin 300192, China
| | - Liu Qin
- Tianjin Research Institute of Water Transport Engineering, M.O.T., Tianjin 300456, China
- National Engineering Research Center of Port Hydraulic Construction Technology, Tianjin 300456, China
| | - Wei Wang
- Tianjin Research Institute of Water Transport Engineering, M.O.T., Tianjin 300456, China
- National Engineering Research Center of Port Hydraulic Construction Technology, Tianjin 300456, China
| |
Collapse
|
12
|
Furuta K, Tang X, Islam S, Tapia A, Chen ZB, Ibrahim SH. Endotheliopathy in the metabolic syndrome: Mechanisms and clinical implications. Pharmacol Ther 2023; 244:108372. [PMID: 36894027 PMCID: PMC10084912 DOI: 10.1016/j.pharmthera.2023.108372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/25/2023] [Accepted: 02/28/2023] [Indexed: 03/09/2023]
Abstract
The increasing prevalence of the metabolic syndrome (MetS) is a threat to global public health due to its lethal complications. Nonalcoholic fatty liver disease (NAFLD) is the hepatic manifestation of the MetS characterized by hepatic steatosis, which is potentially progressive to the inflammatory and fibrotic nonalcoholic steatohepatitis (NASH). The adipose tissue (AT) is also a major metabolic organ responsible for the regulation of whole-body energy homeostasis, and thereby highly involved in the pathogenesis of the MetS. Recent studies suggest that endothelial cells (ECs) in the liver and AT are not just inert conduits but also crucial mediators in various biological processes via the interaction with other cell types in the microenvironment both under physiological and pathological conditions. Herein, we highlight the current knowledge of the role of the specialized liver sinusoidal endothelial cells (LSECs) in NAFLD pathophysiology. Next, we discuss the processes through which AT EC dysfunction leads to MetS progression, with a focus on inflammation and angiogenesis in the AT as well as on endothelial-to-mesenchymal transition of AT-ECs. In addition, we touch upon the function of ECs residing in other metabolic organs including the pancreatic islet and the gut, the dysregulation of which may also contribute to the MetS. Finally, we highlight potential EC-based therapeutic targets for human MetS, and NASH based on recent achievements in basic and clinical research and discuss how to approach unsolved problems in the field.
Collapse
Affiliation(s)
- Kunimaro Furuta
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN, USA; Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Xiaofang Tang
- Department of Diabetes Complications & Metabolism, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Shahidul Islam
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Alonso Tapia
- Department of Diabetes Complications & Metabolism, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Zhen Bouman Chen
- Department of Diabetes Complications & Metabolism, City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| | - Samar H Ibrahim
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN, USA; Division of Pediatric Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
13
|
Sabaté Del Río J, Ro J, Yoon H, Park TE, Cho YK. Integrated technologies for continuous monitoring of organs-on-chips: Current challenges and potential solutions. Biosens Bioelectron 2023; 224:115057. [PMID: 36640548 DOI: 10.1016/j.bios.2022.115057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 12/29/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023]
Abstract
Organs-on-chips (OoCs) are biomimetic in vitro systems based on microfluidic cell cultures that recapitulate the in vivo physicochemical microenvironments and the physiologies and key functional units of specific human organs. These systems are versatile and can be customized to investigate organ-specific physiology, pathology, or pharmacology. They are more physiologically relevant than traditional two-dimensional cultures, can potentially replace the animal models or reduce the use of these models, and represent a unique opportunity for the development of personalized medicine when combined with human induced pluripotent stem cells. Continuous monitoring of important quality parameters of OoCs via a label-free, non-destructive, reliable, high-throughput, and multiplex method is critical for assessing the conditions of these systems and generating relevant analytical data; moreover, elaboration of quality predictive models is required for clinical trials of OoCs. Presently, these analytical data are obtained by manual or automatic sampling and analyzed using single-point, off-chip traditional methods. In this review, we describe recent efforts to integrate biosensing technologies into OoCs for monitoring the physiologies, functions, and physicochemical microenvironments of OoCs. Furthermore, we present potential alternative solutions to current challenges and future directions for the application of artificial intelligence in the development of OoCs and cyber-physical systems. These "smart" OoCs can learn and make autonomous decisions for process optimization, self-regulation, and data analysis.
Collapse
Affiliation(s)
- Jonathan Sabaté Del Río
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, 44919, Republic of Korea
| | - Jooyoung Ro
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, 44919, Republic of Korea; Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Heejeong Yoon
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Tae-Eun Park
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea.
| | - Yoon-Kyoung Cho
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, 44919, Republic of Korea; Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea.
| |
Collapse
|
14
|
Su SH, Song Y, Stephens A, Situ M, McCloskey MC, McGrath JL, Andjelkovic AV, Singer BH, Kurabayashi K. A tissue chip with integrated digital immunosensors: In situ brain endothelial barrier cytokine secretion monitoring. Biosens Bioelectron 2023; 224:115030. [PMID: 36603283 PMCID: PMC10401069 DOI: 10.1016/j.bios.2022.115030] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/08/2022] [Accepted: 12/19/2022] [Indexed: 12/25/2022]
Abstract
Organ-on-a-chip platforms have potential to offer more cost-effective, ethical, and human-resembling models than animal models for disease study and drug discovery. Particularly, the Blood-Brain-Barrier-on-a-chip (BBB-oC) has emerged as a promising tool to investigate several neurological disorders since it promises to provide a model of the multifunctional tissue working as an important node to control pathogen entry, drug delivery and neuroinflammation. A comprehensive understanding of the multiple physiological functions of the tissue model requires biosensors detecting several tissue-secreted substances in a BBB-oC system. However, current sensor-integrated BBB-oC platforms are only available for tissue membrane integrity characterization based on permeability measurement. Protein secretory pathways are closely associated with the tissue's various diseased conditions. At present, no biosensor-integrated BBB-oC platform exists that permits in situ tissue protein secretion analysis over time, which prohibits researchers from fully understanding the time-evolving pathology of a tissue barrier. Herein, the authors present a platform named "Digital Tissue-BArrier-CytoKine-counting-on-a-chip (DigiTACK)," which integrates digital immunosensors into a tissue chip system and demonstrates on-chip multiplexed, ultrasensitive, longitudinal cytokine secretion profiling of cultured brain endothelial barrier tissues. The integrated digital sensors utilize a novel beadless microwell format to perform an ultrafast "digital fingerprinting" of the analytes while achieving a low limit of detection (LoD) around 100-500 fg/mL for mouse MCP1 (CCL2), IL-6 and KC (CXCL1). The DigiTACK platform is extensively applicable to profile temporal cytokine secretion of other barrier-related organ-on-a-chip systems and can provide new insight into the secretory dynamics of the BBB by sequentially controlled experiments.
Collapse
Affiliation(s)
- Shiuan-Haur Su
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yujing Song
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Andrew Stephens
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Muyu Situ
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Molly C McCloskey
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - James L McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Anuska V Andjelkovic
- Department of Pathology and Neurosurgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Benjamin H Singer
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, 48109, USA; Weil Institute for Critical Care Research and Innovation, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Katsuo Kurabayashi
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, 48109, USA; Weil Institute for Critical Care Research and Innovation, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
15
|
Simone G. Trends of Biosensing: Plasmonics through Miniaturization and Quantum Sensing. Crit Rev Anal Chem 2023; 54:2183-2208. [PMID: 36601882 DOI: 10.1080/10408347.2022.2161813] [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] [Indexed: 01/06/2023]
Abstract
Despite being extremely old concepts, plasmonics and surface plasmon resonance-based biosensors have been increasingly popular in the recent two decades due to the growing interest in nanooptics and are now of relevant significance in regards to applications associated with human health. Plasmonics integration into point-of-care devices for health surveillance has enabled significant levels of sensitivity and limit of detection to be achieved and has encouraged the expansion of the fields of study and market niches devoted to the creation of quick and incredibly sensitive label-free detection. The trend reflects in wearable plasmonic sensor development as well as point-of-care applications for widespread applications, demonstrating the potential impact of the new generation of plasmonic biosensors on human well-being through the concepts of personalized medicine and global health. In this context, the aim here is to discuss the potential, limitations, and opportunities for improvement that have arisen as a result of the integration of plasmonics into microsystems and lab-on-chip over the past five years. Recent applications of plasmonic biosensors in microsystems and sensor performance are analyzed. The final analysis focuses on the integration of microfluidics and lab-on-a-chip with quantum plasmonics technology prospecting it as a promising solution for chemical and biological sensing. Here it is underlined how the research in the field of quantum plasmonic sensing for biological applications has flourished over the past decade with the aim to overcome the limits given by quantum fluctuations and noise. The significant advances in nanophotonics, plasmonics and microsystems used to create increasingly effective biosensors would continue to benefit this field if harnessed properly.
Collapse
Affiliation(s)
- Giuseppina Simone
- Chemical Engineering, University of Naples 'Federico II', Naples, Italy
| |
Collapse
|
16
|
Hassanzadeh P, Atyabi F, Dinarvand R. Technical and engineering considerations for designing therapeutics and delivery systems. J Control Release 2023; 353:411-422. [PMID: 36470331 DOI: 10.1016/j.jconrel.2022.11.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
The newly-emerged pathological conditions and increased rates of drug resistance necessitate application of the state-of-the-art technologies for accelerated discovery of the therapeutic candidates and obtaining comprehensive knowledge about their targets, action mechanisms, and interactions within the body including those between the receptors and drugs. Using the physics- and chemistry-based modern techniques for theranostic purposes, preparing smart carriers, local delivery of genes or drugs, and enhancing pharmaceutical bioavailability could be of great value against the hard-to-treat diseases and growing drug resistance. Besides the artificial intelligence- and quantum-based techniques, crystal engineering capable of designing new molecules with appropriate characteristics, improving the stability and bioavailability of poorly soluble drugs, and efficient carrier development could play a crucial role in manufacturing efficient pharmaceuticals and reducing the adverse events. In this context, identifying the structures and behaviors of crystals and predicting their characteristics are of great value. Electron diffraction by accelerated analysis of the chemicals and sensitivity to charge alterations, electromechanical tools for controlled delivery of therapeutics, mechatronics via fabrication of multi-functional smart products including the organ-on-chip devices for healthcare applications, and optomechatronics by overcoming the limitations of conventional biomedical techniques could address the unmet biomedical requirements and facilitate development of more effective theranostics with improved outcomes.
Collapse
Affiliation(s)
- Parichehr Hassanzadeh
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 13169-43551, Iran; Sasan Hospital, Tehran 14159-83391, Iran.
| | - Fatemeh Atyabi
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 13169-43551, Iran
| | - Rassoul Dinarvand
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 13169-43551, Iran
| |
Collapse
|
17
|
Victorious A. Current Applications of Organ-on-a-Chip: A Step Closer to Personalized Medicine. BIO INTEGRATION 2022. [DOI: 10.15212/bioi-2022-0027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Abstract In the pharmaceutical industry, a critical need exists for effective drug development approaches that better account for factors imposed by the physiological microenvironment. Organ-on-a-chip (OOAC)—a revolutionary technology that simulates human organs’
physiological milieu and performance on a chip—has applications in curing illnesses and drug screening, and enormous potential to transform the drug discovery workflow. However, the effective integration of this unique engineering system into ordinary pharmacological and medical contexts
remains in development. This Editorial summarizes current research on OOAC systems, and offers insight into future development prospects and the need for a next-generation OOAC framework.
Collapse
|
18
|
Ultrasensitive rapid cytokine sensors based on asymmetric geometry two-dimensional MoS 2 diodes. Nat Commun 2022; 13:7593. [PMID: 36535944 PMCID: PMC9763493 DOI: 10.1038/s41467-022-35278-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 11/24/2022] [Indexed: 12/23/2022] Open
Abstract
The elevation of cytokine levels in body fluids has been associated with numerous health conditions. The detection of these cytokine biomarkers at low concentrations may help clinicians diagnose diseases at an early stage. Here, we report an asymmetric geometry MoS2 diode-based biosensor for rapid, label-free, highly sensitive, and specific detection of tumor necrosis factor-α (TNF-α), a proinflammatory cytokine. This sensor is functionalized with TNF-α binding aptamers to detect TNF-α at concentrations as low as 10 fM, well below the typical concentrations found in healthy blood. Interactions between aptamers and TNF-α at the sensor surface induce a change in surface energy that alters the current-voltage rectification behavior of the MoS2 diode, which can be read out using a two-electrode configuration. The key advantages of this diode sensor are the simple fabrication process and electrical readout, and therefore, the potential to be applied in a rapid and easy-to-use, point-of-care, diagnostic tool.
Collapse
|
19
|
He J, Zhou L, Huang G, Shen J, Chen W, Wang C, Kim A, Zhang Z, Cheng W, Dai S, Ding F, Chen P. Enhanced Label-Free Nanoplasmonic Cytokine Detection in SARS-CoV-2 Induced Inflammation Using Rationally Designed Peptide Aptamer. ACS APPLIED MATERIALS & INTERFACES 2022; 14:48464-48475. [PMID: 36281943 PMCID: PMC9627400 DOI: 10.1021/acsami.2c14748] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/13/2022] [Indexed: 06/12/2023]
Abstract
Rapid and precise serum cytokine quantification provides immense clinical significance in monitoring the immune status of patients in rapidly evolving infectious/inflammatory disorders, examplified by the ongoing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic. However, real-time information on predictive cytokine biomarkers to guide targetable immune pathways in pathogenic inflammation is critically lacking, because of the insufficient detection range and detection limit in current label-free cytokine immunoassays. In this work, we report a highly sensitive localized surface plasmon resonance imaging (LSPRi) immunoassay for label-free Interleukin 6 (IL-6) detection utilizing rationally designed peptide aptamers as the capture interface. Benefiting from its characteristically smaller dimension and direct functionalization on the sensing surface via Au-S bonding, the peptide-aptamer-based LSPRi immunoassay achieved enhanced label-free serum IL-6 detection with a record-breaking limit of detection down to 4.6 pg/mL, and a wide dynamic range of ∼6 orders of magnitude (values from 4.6 to 1 × 106 pg/mL were observed). The immunoassay was validated in vitro for label-free analysis of SARS-CoV-2 induced inflammation, and further applied in rapid quantification of serum IL-6 profiles in COVID-19 patients. Our peptide aptamer LSPRi immunoassay demonstrates great potency in label-free cytokine detection with unprecedented sensing capability to provide accurate and timely interpretation of the inflammatory status and disease progression, and determination of prognosis.
Collapse
Affiliation(s)
- Jiacheng He
- Materials Research and Education Center, Auburn University, Auburn, Alabama36849, United States
| | - Lang Zhou
- Materials Research and Education Center, Auburn University, Auburn, Alabama36849, United States
| | - Gangtong Huang
- Department of Physics and Astronomy, Clemson University, Clemson, South Carolina29634, United States
| | - Jialiang Shen
- Materials Research and Education Center, Auburn University, Auburn, Alabama36849, United States
| | - Wu Chen
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, Alabama36849, United States
| | - Chuanyu Wang
- Materials Research and Education Center, Auburn University, Auburn, Alabama36849, United States
| | - Albert Kim
- Center for Medicine, Health, and Society, Vanderbilt University, Nashville, Tennessee37235, United States
| | - Zhuoyu Zhang
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, New York11201, United States
- Department of Biomedical Engineering, New York University, Brooklyn, New York11201, United States
| | - Weiqiang Cheng
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, New York11201, United States
- Department of Biomedical Engineering, New York University, Brooklyn, New York11201, United States
| | - Siyuan Dai
- Materials Research and Education Center, Auburn University, Auburn, Alabama36849, United States
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, South Carolina29634, United States
| | - Pengyu Chen
- Materials Research and Education Center, Auburn University, Auburn, Alabama36849, United States
| |
Collapse
|
20
|
Mou L, Mandal K, Mecwan MM, Hernandez AL, Maity S, Sharma S, Herculano RD, Kawakita S, Jucaud V, Dokmeci MR, Khademhosseini A. Integrated biosensors for monitoring microphysiological systems. LAB ON A CHIP 2022; 22:3801-3816. [PMID: 36074812 PMCID: PMC9635816 DOI: 10.1039/d2lc00262k] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Microphysiological systems (MPSs), also known as organ-on-a-chip models, aim to recapitulate the functional components of human tissues or organs in vitro. Over the last decade, with the advances in biomaterials, 3D bioprinting, and microfluidics, numerous MPSs have emerged with applications to study diseased and healthy tissue models. Various organs have been modeled using MPS technology, such as the heart, liver, lung, and blood-brain barrier. An important aspect of in vitro modeling is the accurate phenotypical and functional characterization of the modeled organ. However, most conventional characterization methods are invasive and destructive and do not allow continuous monitoring of the cells in culture. On the other hand, microfluidic biosensors enable in-line, real-time sensing of target molecules with an excellent limit of detection and in a non-invasive manner, thereby effectively overcoming the limitation of the traditional techniques. Consequently, microfluidic biosensors have been increasingly integrated into MPSs and used for in-line target detection. This review discusses the state-of-the-art microfluidic biosensors by providing specific examples, detailing their main advantages in monitoring MPSs, and highlighting current developments in this field. Finally, we describe the remaining challenges and potential future developments to advance the current state-of-the-art in integrated microfluidic biosensors.
Collapse
Affiliation(s)
- Lei Mou
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, California, USA.
- Department of Clinical Laboratory, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, No. 63 Duobao Road, Liwan District, Guangzhou, Guangdong, P. R. China
| | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, California, USA.
| | - Marvin Magan Mecwan
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, California, USA.
| | - Ana Lopez Hernandez
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, California, USA.
| | - Surjendu Maity
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, California, USA.
| | - Saurabh Sharma
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, California, USA.
| | - Rondinelli Donizetti Herculano
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, California, USA.
- Department of Bioprocess and Biotechnology Engineering, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP 14801-902, Brazil
| | - Satoru Kawakita
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, California, USA.
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, California, USA.
| | - Mehmet Remzi Dokmeci
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, California, USA.
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, California, USA.
| |
Collapse
|
21
|
Das CM, Kong KV, Yong KT. Diagnostic plasmonic sensors: opportunities and challenges. Chem Commun (Camb) 2022; 58:9573-9585. [PMID: 35975603 DOI: 10.1039/d2cc03431j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The medical fraternity is currently burgeoned and stressed with a huge rush of patients who have inflammatory conditions, metabolite diseases, and cardiovascular diseases. In these circumstances, advanced sensing technologies could have a huge impact on the quality of life of patients. Given plasmonic resonance effects significantly improve the ability to rapidly and accurately detect biological markers, plasmonic technology is harnessed to develop a fast and accurate diagnosis that can provide timely intervention with the diseases and can also aid the recovery process by complementing the therapy stage. In this short review, we provide an overlook of how the field of plasmonic sensing has revolutionized the field of medical diagnostics. This article reviews the fundamentals and development of plasmonics. In addition, we highlight the sensitivity of various SPR and LSPR sensors. The chemistry for functionalizing plasmonic sensors is also discussed. This review also outlines some general suggestions for future directions that we feel might be useful to advance our understanding of the universe or speed up the development of plasmonic sensors in the future.
Collapse
Affiliation(s)
- Chandreyee Manas Das
- School of Electrical and Electronic Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore.,CINTRA CNRS/NTU/THALES, UMI 3288, Research Techno Plaza, 50 Nanyang Drive, Border X Block, 637553, Singapore
| | - Kien Voon Kong
- Department of Chemistry, National Taiwan University, Taipei City, Taiwan
| | - Ken-Tye Yong
- School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia. .,The University of Sydney Nano Institute, The University of Sydney, Sydney, New South Wales 2006, Australia.,The Biophotonics and MechanoBioengineering Lab, The University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
22
|
Rogal J, Roosz J, Teufel C, Cipriano M, Xu R, Eisler W, Weiss M, Schenke‐Layland K, Loskill P. Autologous Human Immunocompetent White Adipose Tissue-on-Chip. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104451. [PMID: 35466539 PMCID: PMC9218765 DOI: 10.1002/advs.202104451] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 03/03/2022] [Indexed: 05/07/2023]
Abstract
Obesity and associated diseases, such as diabetes, have reached epidemic proportions globally. In this era of "diabesity", white adipose tissue (WAT) has become a target of high interest for therapeutic strategies. To gain insights into mechanisms of adipose (patho-)physiology, researchers traditionally relied on animal models. Leveraging Organ-on-Chip technology, a microphysiological in vitro model of human WAT is introduced: a tailored microfluidic platform featuring vasculature-like perfusion that integrates 3D tissues comprising all major WAT-associated cellular components (mature adipocytes, organotypic endothelial barriers, stromovascular cells including adipose tissue macrophages) in an autologous manner and recapitulates pivotal WAT functions, such as energy storage and mobilization as well as endocrine and immunomodulatory activities. A precisely controllable bottom-up approach enables the generation of a multitude of replicates per donor circumventing inter-donor variability issues and paving the way for personalized medicine. Moreover, it allows to adjust the model's degree of complexity via a flexible mix-and-match approach. This WAT-on-Chip system constitutes the first human-based, autologous, and immunocompetent in vitro adipose tissue model that recapitulates almost full tissue heterogeneity and can become a powerful tool for human-relevant research in the field of metabolism and its associated diseases as well as for compound testing and personalized- and precision medicine applications.
Collapse
Affiliation(s)
- Julia Rogal
- Department for Microphysiological Systems, Institute of Biomedical EngineeringEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGBNobelstr. 12Stuttgart70569Germany
| | - Julia Roosz
- NMI Natural and Medical Sciences Institute at the University of TübingenMarkwiesenstr. 55Reutlingen72770Germany
| | - Claudia Teufel
- Department for Microphysiological Systems, Institute of Biomedical EngineeringEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
| | - Madalena Cipriano
- Department for Microphysiological Systems, Institute of Biomedical EngineeringEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
- 3R‐Center for In vitro Models and Alternatives to Animal TestingEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
| | - Raylin Xu
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGBNobelstr. 12Stuttgart70569Germany
- Harvard Medical School (HMS)25 Shattuck StBostonMA02115USA
| | - Wiebke Eisler
- Clinic for PlasticReconstructiveHand and Burn SurgeryBG Trauma CenterEberhard Karls University TübingenSchnarrenbergstraße 95Tübingen72076Germany
| | - Martin Weiss
- NMI Natural and Medical Sciences Institute at the University of TübingenMarkwiesenstr. 55Reutlingen72770Germany
- Department of Women's HealthEberhard Karls University TübingenCalwerstrasse 7Tübingen72076Germany
| | - Katja Schenke‐Layland
- NMI Natural and Medical Sciences Institute at the University of TübingenMarkwiesenstr. 55Reutlingen72770Germany
- Department of Medicine/CardiologyCardiovascular Research LaboratoriesDavid Geffen School of Medicine at UCLA675 Charles E. Young Drive South, MRL 3645Los AngelesCA90095USA
- Cluster of Excellence iFIT (EXC2180) “Image‐Guided and Functionally Instructed Tumor Therapies”Eberhard Karls University TuebingenRöntgenweg 11Tuebingen72076Germany
- Department for Medical Technologies and Regenerative MedicineInstitute of Biomedical EngineeringEberhard Karls University TübingenSilcherstr. 7/1Tübingen72076Germany
| | - Peter Loskill
- Department for Microphysiological Systems, Institute of Biomedical EngineeringEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
- NMI Natural and Medical Sciences Institute at the University of TübingenMarkwiesenstr. 55Reutlingen72770Germany
- 3R‐Center for In vitro Models and Alternatives to Animal TestingEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
| |
Collapse
|
23
|
Kavand H, Nasiri R, Herland A. Advanced Materials and Sensors for Microphysiological Systems: Focus on Electronic and Electrooptical Interfaces. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107876. [PMID: 34913206 DOI: 10.1002/adma.202107876] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/07/2021] [Indexed: 06/14/2023]
Abstract
Advanced in vitro cell culture systems or microphysiological systems (MPSs), including microfluidic organ-on-a-chip (OoC), are breakthrough technologies in biomedicine. These systems recapitulate features of human tissues outside of the body. They are increasingly being used to study the functionality of different organs for applications such as drug evolutions, disease modeling, and precision medicine. Currently, developers and endpoint users of these in vitro models promote how they can replace animal models or even be a better ethically neutral and humanized alternative to study pathology, physiology, and pharmacology. Although reported models show a remarkable physiological structure and function compared to the conventional 2D cell culture, they are almost exclusively based on standard passive polymers or glass with none or minimal real-time stimuli and readout capacity. The next technology leap in reproducing in vivo-like functionality and real-time monitoring of tissue function could be realized with advanced functional materials and devices. This review describes the currently reported electronic and optical advanced materials for sensing and stimulation of MPS models. In addition, an overview of multi-sensing for Body-on-Chip platforms is given. Finally, one gives the perspective on how advanced functional materials could be integrated into in vitro systems to precisely mimic human physiology.
Collapse
Affiliation(s)
- Hanie Kavand
- Division of Micro- and Nanosystems, Department of Intelligent Systems, KTH Royal Institute of Technology, Malvinas Väg 10 pl 5, Stockholm, 100 44, Sweden
| | - Rohollah Nasiri
- AIMES, Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, Solnavägen 9/B8, Solna, 171 65, Sweden
- Division of Nanobiotechnology, Department of Protein Science, KTH Royal Institute of Technology, Tomtebodavägen 23a, Solna, 171 65, Sweden
| | - Anna Herland
- Division of Micro- and Nanosystems, Department of Intelligent Systems, KTH Royal Institute of Technology, Malvinas Väg 10 pl 5, Stockholm, 100 44, Sweden
- AIMES, Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, Solnavägen 9/B8, Solna, 171 65, Sweden
- Division of Nanobiotechnology, Department of Protein Science, KTH Royal Institute of Technology, Tomtebodavägen 23a, Solna, 171 65, Sweden
| |
Collapse
|
24
|
Alba-Patiño A, Vaquer A, Barón E, Russell SM, Borges M, de la Rica R. Micro- and nanosensors for detecting blood pathogens and biomarkers at different points of sepsis care. Mikrochim Acta 2022; 189:74. [PMID: 35080669 PMCID: PMC8790942 DOI: 10.1007/s00604-022-05171-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/26/2021] [Indexed: 12/29/2022]
Abstract
Severe infections can cause a dysregulated response leading to organ dysfunction known as sepsis. Sepsis can be lethal if not identified and treated right away. This requires measuring biomarkers and pathogens rapidly at the different points where sepsis care is provided. Current commercial approaches for sepsis diagnosis are not fast, sensitive, and/or specific enough for meeting this medical challenge. In this article, we review recent advances in the development of diagnostic tools for sepsis management based on micro- and nanostructured materials. We start with a brief introduction to the most popular biomarkers for sepsis diagnosis (lactate, procalcitonin, cytokines, C-reactive protein, and other emerging protein and non-protein biomarkers including miRNAs and cell-based assays) and methods for detecting bacteremia. We then highlight the role of nano- and microstructured materials in developing biosensors for detecting them taking into consideration the particular needs of every point of sepsis care (e.g., ultrafast detection of multiple protein biomarkers for diagnosing in triage, emergency room, ward, and intensive care unit; quantitative detection to de-escalate treatment; ultrasensitive and culture-independent detection of blood pathogens for personalized antimicrobial therapies; robust, portable, and web-connected biomarker tests outside the hospital). We conclude with an overview of the most utilized nano- and microstructured materials used thus far for solving issues related to sepsis diagnosis and point to new challenges for future development.
Collapse
Affiliation(s)
- Alejandra Alba-Patiño
- Multidisciplinary Sepsis Group, Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Department of Chemistry, University of the Balearic Islands, Palma, Spain
| | - Andreu Vaquer
- Multidisciplinary Sepsis Group, Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Department of Chemistry, University of the Balearic Islands, Palma, Spain
| | - Enrique Barón
- Multidisciplinary Sepsis Group, Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.
| | - Steven M Russell
- Multidisciplinary Sepsis Group, Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Marcio Borges
- Multidisciplinary Sepsis Group, Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Multidisciplinary Sepsis Unit, ICU, Son Llàtzer University Hospital, Palma, Spain
| | - Roberto de la Rica
- Multidisciplinary Sepsis Group, Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.
| |
Collapse
|
25
|
Joint-on-chip platforms: entering a new era of in vitro models for arthritis. Nat Rev Rheumatol 2022; 18:217-231. [PMID: 35058618 DOI: 10.1038/s41584-021-00736-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2021] [Indexed: 12/12/2022]
|
26
|
Qi L, Zushin PJ, Chang CF, Lee YT, Alba DL, Koliwad S, Stahl A. Probing Insulin Sensitivity with Metabolically Competent Human Stem Cell-Derived White Adipose Tissue Microphysiological Systems. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2103157. [PMID: 34761526 PMCID: PMC8776615 DOI: 10.1002/smll.202103157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/21/2021] [Indexed: 05/13/2023]
Abstract
Impaired white adipose tissue (WAT) function has been recognized as a critical early event in obesity-driven disorders, but high buoyancy, fragility, and heterogeneity of primary adipocytes have largely prevented their use in drug discovery efforts highlighting the need for human stem cell-based approaches. Here, human stem cells are utilized to derive metabolically functional 3D adipose tissue (iADIPO) in a microphysiological system (MPS). Surprisingly, previously reported WAT differentiation approaches create insulin resistant WAT ill-suited for type-2 diabetes mellitus drug discovery. Using three independent insulin sensitivity assays, i.e., glucose and fatty acid uptake and suppression of lipolysis, as the functional readouts new differentiation conditions yielding hormonally responsive iADIPO are derived. Through concomitant optimization of an iADIPO-MPS, it is abled to obtain WAT with more unilocular and significantly larger (≈40%) lipid droplets compared to iADIPO in 2D culture, increased insulin responsiveness of glucose uptake (≈2-3 fold), fatty acid uptake (≈3-6 fold), and ≈40% suppressing of stimulated lipolysis giving a dynamic range that is competent to current in vivo and ex vivo models, allowing to identify both insulin sensitizers and desensitizers.
Collapse
Affiliation(s)
- Lin Qi
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California, Berkeley, Berkeley, California, 94720, USA
| | - Peter James Zushin
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California, Berkeley, Berkeley, California, 94720, USA
| | - Ching-Fang Chang
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California, Berkeley, Berkeley, California, 94720, USA
| | - Yue Tung Lee
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California, Berkeley, Berkeley, California, 94720, USA
| | - Diana L. Alba
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of California, San Francisco; Diabetes Center, University of California, San Francisco, San Francisco, California 94143, USA
| | - Suneil Koliwad
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of California, San Francisco; Diabetes Center, University of California, San Francisco, San Francisco, California 94143, USA
| | - Andreas Stahl
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California, Berkeley, Berkeley, California, 94720, USA
| |
Collapse
|
27
|
Kim J, Noh S, Park JA, Park SC, Park SJ, Lee JH, Ahn JH, Lee T. Recent Advances in Aptasensor for Cytokine Detection: A Review. SENSORS (BASEL, SWITZERLAND) 2021; 21:8491. [PMID: 34960590 PMCID: PMC8705356 DOI: 10.3390/s21248491] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/03/2021] [Accepted: 12/12/2021] [Indexed: 12/14/2022]
Abstract
Cytokines are proteins secreted by immune cells. They promote cell signal transduction and are involved in cell replication, death, and recovery. Cytokines are immune modulators, but their excessive secretion causes uncontrolled inflammation that attacks normal cells. Considering the properties of cytokines, monitoring the secretion of cytokines in vivo is of great value for medical and biological research. In this review, we offer a report on recent studies for cytokine detection, especially studies on aptasensors using aptamers. Aptamers are single strand nucleic acids that form a stable three-dimensional structure and have been receiving attention due to various characteristics such as simple production methods, low molecular weight, and ease of modification while performing a physiological role similar to antibodies.
Collapse
Affiliation(s)
- Jinmyeong Kim
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-Ro, Nowon-gu, Seoul 01897, Korea; (J.K.); (S.N.); (J.A.P.)
| | - Seungwoo Noh
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-Ro, Nowon-gu, Seoul 01897, Korea; (J.K.); (S.N.); (J.A.P.)
| | - Jeong Ah Park
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-Ro, Nowon-gu, Seoul 01897, Korea; (J.K.); (S.N.); (J.A.P.)
| | - Sang-Chan Park
- Department of Electronics Engineering, Chungnam National University, 99 Yuseong-gu, Daejeon 34134, Korea;
| | - Seong Jun Park
- Department of Electrical Engineering, Kwangwoon University, 20 Kwangwoon-Ro, Nowon-gu, Seoul 01897, Korea;
| | - Jin-Ho Lee
- School of Biomedical Convergence Engineering, Pusan National University, 49 Busandaehak-ro, Yangsan 50612, Korea;
| | - Jae-Hyuk Ahn
- Department of Electronics Engineering, Chungnam National University, 99 Yuseong-gu, Daejeon 34134, Korea;
| | - Taek Lee
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-Ro, Nowon-gu, Seoul 01897, Korea; (J.K.); (S.N.); (J.A.P.)
| |
Collapse
|
28
|
Wang C, Huang CH, Gao Z, Shen J, He J, MacLachlan A, Ma C, Chang Y, Yang W, Cai Y, Lou Y, Dai S, Chen W, Li F, Chen P. Nanoplasmonic Sandwich Immunoassay for Tumor-Derived Exosome Detection and Exosomal PD-L1 Profiling. ACS Sens 2021; 6:3308-3319. [PMID: 34494426 DOI: 10.1021/acssensors.1c01101] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumor-derived exosomes play a vital role in the process of cancer development. Quantitative analysis of exosomes and exosome-shuttled proteins would be of immense value in understanding cancer progression and generating reliable predictive biomarkers for cancer diagnosis and treatment. Recent studies have indicated the critical role of exosomal programmed death ligand 1 (PD-L1) in immune checkpoint therapy and its application as a patient stratification biomarker in cancer immunotherapy. Here, we present a nanoplasmonic exosome immunoassay utilizing gold-silver (Au@Ag) core-shell nanobipyramids and gold nanorods, which form sandwich immune complexes with target exosomes. The immunoassay generates a distinct plasmonic signal pattern unique to exosomes with specific exosomal PD-L1 expression, allowing rapid, highly sensitive exosome detection and accurate identification of PD-L1 exosome subtypes in a single assay. The developed nanoplasmonic sandwich immunoassay provides a novel and viable approach for tumor cell-derived exosome detection and analysis with quantitative molecular details of key exosomal proteins, manifesting its great potential as a transformative diagnostic tool for early cancer detection, prognosis, and post-treatment monitoring.
Collapse
Affiliation(s)
- Chuanyu Wang
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, Alabama 36849, United States
| | - Chung-Hui Huang
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, Alabama 36849, United States
| | - Zhuangqiang Gao
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, Alabama 36849, United States
| | - Jialiang Shen
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, Alabama 36849, United States
| | - Jiacheng He
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, Alabama 36849, United States
| | - Alana MacLachlan
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, Alabama 36849, United States
| | - Chao Ma
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, New York 11201, United States
- Department of Biomedical Engineering, New York University, Brooklyn, New York 11201, United States
| | - Ya Chang
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, Alabama 36849, United States
| | - Wen Yang
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, Alabama 36849, United States
| | - Yuxin Cai
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, Alabama 36849, United States
| | - Yang Lou
- Yurogen Biosystems LLC, Worcester, Massachusetts 01605, United States
| | - Siyuan Dai
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, Alabama 36849, United States
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, New York 11201, United States
- Department of Biomedical Engineering, New York University, Brooklyn, New York 11201, United States
| | - Feng Li
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, Alabama 36849, United States
| | - Pengyu Chen
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, Alabama 36849, United States
| |
Collapse
|
29
|
Liu C, Chu D, Kalantar‐Zadeh K, George J, Young HA, Liu G. Cytokines: From Clinical Significance to Quantification. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2004433. [PMID: 34114369 PMCID: PMC8336501 DOI: 10.1002/advs.202004433] [Citation(s) in RCA: 301] [Impact Index Per Article: 75.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/26/2021] [Indexed: 05/24/2023]
Abstract
Cytokines are critical mediators that oversee and regulate immune and inflammatory responses via complex networks and serve as biomarkers for many diseases. Quantification of cytokines has significant value in both clinical medicine and biology as the levels provide insights into physiological and pathological processes and can be used to aid diagnosis and treatment. Cytokines and their clinical significance are introduced from the perspective of their pro- and anti-inflammatory effects. Factors affecting cytokines quantification in biological fluids, native levels in different body fluids, sample processing and storage conditions, sensitivity to freeze-thaw, and soluble cytokine receptors are discussed. In addition, recent advances in in vitro and in vivo assays, biosensors based on different signal outputs and intracellular to extracellular protein expression are summarized. Various quantification platforms for high-sensitivity and reliable measurement of cytokines in different scenarios are discussed, and commercially available cytokine assays are compared. A discussion of challenges in the development and advancement of technologies for cytokine quantification that aim to achieve real-time multiplex cytokine analysis for point-of-care situations applicable for both biomedical research and clinical practice are discussed.
Collapse
Affiliation(s)
- Chao Liu
- School of Materials Science and EngineeringUniversity of New South WalesSydneyNSW2052Australia
| | - Dewei Chu
- School of Materials Science and EngineeringUniversity of New South WalesSydneyNSW2052Australia
| | | | - Jacob George
- Storr Liver CentreWestmead Institute of Medical ResearchUniversity of Sydney and Department of Gastroenterology and HepatologyWestmead HospitalWestmeadNSW2145Australia
| | - Howard A. Young
- Laboratory of Cancer ImmunometabolismCenter for Cancer ResearchNational Cancer Institute at FrederickFrederickMD21702USA
| | - Guozhen Liu
- School of Life and Health SciencesThe Chinese University of Hong KongShenzhen518172P. R. China
- Graduate School of Biomedical EngineeringUniversity of New South WalesSydneyNSW2052Australia
| |
Collapse
|
30
|
Su SH, Song Y, Newstead MW, Cai T, Wu M, Stephens A, Singer BH, Kurabayashi K. Ultrasensitive Multiparameter Phenotyping of Rare Cells Using an Integrated Digital-Molecular-Counting Microfluidic Well Plate. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2101743. [PMID: 34170616 PMCID: PMC8349899 DOI: 10.1002/smll.202101743] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/08/2021] [Indexed: 06/13/2023]
Abstract
Integrated microfluidic cellular phenotyping platforms provide a promising means of studying a variety of inflammatory diseases mediated by cell-secreted cytokines. However, immunosensors integrated in previous microfluidic platforms lack the sensitivity to detect small signals in the cellular secretion of proinflammatory cytokines with high precision. This limitation prohibits researchers from studying cells secreting cytokines at low abundance or existing at a small population. Herein, the authors present an integrated platform named the "digital Phenoplate (dPP)," which integrates digital immunosensors into a microfluidic chip with on-chip cell assay chambers, and demonstrates ultrasensitive cellular cytokine secretory profile measurement. The integrated sensors yield a limit of detection as small as 0.25 pg mL-1 for mouse tumor necrosis factor alpha (TNF-α). Each on-chip cell assay chamber confines cells whose population ranges from ≈20 to 600 in arrayed single-cell trapping microwells. Together, these microfluidic features of the dPP simultaneously permit precise counting and image-based cytometry of individual cells while performing parallel measurements of TNF-α released from rare cells under multiple stimulant conditions for multiple samples. The dPP platform is broadly applicable to the characterization of cellular phenotypes demanding high precision and high throughput.
Collapse
Affiliation(s)
- Shiuan-Haur Su
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yujing Song
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Michael W Newstead
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Tao Cai
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - MengXi Wu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Andrew Stephens
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Benjamin H Singer
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Michigan Center for Integrative Research in Critical Care, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Katsuo Kurabayashi
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Michigan Center for Integrative Research in Critical Care, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
31
|
Chen JS, Chen PF, Lin HTH, Huang NT. A Localized surface plasmon resonance (LSPR) sensor integrated automated microfluidic system for multiplex inflammatory biomarker detection. Analyst 2021; 145:7654-7661. [PMID: 32966364 DOI: 10.1039/d0an01201g] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Inflammation is a complex biological response of the human body to external or internal stimuli, such as invading pathogens, defective cells, or irritating substances. One important indicator of inflammatory conditions or the progress of various diseases, such as cancer, cardiovascular diseases, neurological diseases, connective tissue diseases, sepsis, or Alzheimer's disease, is the concentration level of inflammatory biomarkers, including immunoglobulins, cytokines, and C-reactive protein (CRP). Since inflammatory biomarkers are highly correlated with each other, it is important to measure them simultaneously. To enable continuous and dynamic inflammatory biomarker detection, we utilized localized surface plasmon resonance (LSPR) to perform label-free molecule sensing. Since the LSPR sensing mechanism requires only a small sensing area with simplified optical setup, it can be easily integrated with a microfluidic device. To simplify reagent operation complexity, we developed an automated microfluidic control system to control reagent guiding and switching in the immunoassay with less manual processes and potential operation errors. Our results successfully demonstrated multiplex IgG, TNF-α, and CRP measurement with only 60 μL assay volume and 3.5 h assay time. In each test, 20 sensing spot measurements under four different reagent conditions can be performed. Overall, we envision that the LSPR sensor integrated automated microfluidic control system could perform rapid, multiplex, and multiparallel continuous inflammatory biomarker detection, which would be beneficial for various applications, such as immune status monitoring, diagnosis and prognosis of inflammatory diseases.
Collapse
Affiliation(s)
- Jhih-Siang Chen
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, 10617, Taiwan.
| | | | | | | |
Collapse
|
32
|
Boeri L, Perottoni S, Izzo L, Giordano C, Albani D. Microbiota-Host Immunity Communication in Neurodegenerative Disorders: Bioengineering Challenges for In Vitro Modeling. Adv Healthc Mater 2021; 10:e2002043. [PMID: 33661580 PMCID: PMC11468246 DOI: 10.1002/adhm.202002043] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/01/2021] [Indexed: 12/12/2022]
Abstract
Human microbiota communicates with its host by secreting signaling metabolites, enzymes, or structural components. Its homeostasis strongly influences the modulation of human tissue barriers and immune system. Dysbiosis-induced peripheral immunity response can propagate bacterial and pro-inflammatory signals to the whole body, including the brain. This immune-mediated communication may contribute to several neurodegenerative disorders, as Alzheimer's disease. In fact, neurodegeneration is associated with dysbiosis and neuroinflammation. The interplay between the microbial communities and the brain is complex and bidirectional, and a great deal of interest is emerging to define the exact mechanisms. This review focuses on microbiota-immunity-central nervous system (CNS) communication and shows how gut and oral microbiota populations trigger immune cells, propagating inflammation from the periphery to the cerebral parenchyma, thus contributing to the onset and progression of neurodegeneration. Moreover, an overview of the technological challenges with in vitro modeling of the microbiota-immunity-CNS axis, offering interesting technological hints about the most advanced solutions and current technologies is provided.
Collapse
Affiliation(s)
- Lucia Boeri
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoPiazza Leonardo da Vinci 32Milan20133Italy
| | - Simone Perottoni
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoPiazza Leonardo da Vinci 32Milan20133Italy
| | - Luca Izzo
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoPiazza Leonardo da Vinci 32Milan20133Italy
| | - Carmen Giordano
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoPiazza Leonardo da Vinci 32Milan20133Italy
| | - Diego Albani
- Department of NeuroscienceIstituto di Ricerche Farmacologiche Mario Negri IRCCSvia Mario Negri 2Milan20156Italy
| |
Collapse
|
33
|
The Inflammatory Profile of Obesity and the Role on Pulmonary Bacterial and Viral Infections. Int J Mol Sci 2021; 22:ijms22073456. [PMID: 33810619 PMCID: PMC8037155 DOI: 10.3390/ijms22073456] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 12/16/2022] Open
Abstract
Obesity is a globally increasing health problem, entailing diverse comorbidities such as infectious diseases. An obese weight status has marked effects on lung function that can be attributed to mechanical dysfunctions. Moreover, the alterations of adipocyte-derived signal mediators strongly influence the regulation of inflammation, resulting in chronic low-grade inflammation. Our review summarizes the known effects regarding pulmonary bacterial and viral infections. For this, we discuss model systems that allow mechanistic investigation of the interplay between obesity and lung infections. Overall, obesity gives rise to a higher susceptibility to infectious pathogens, but the pathogenetic process is not clearly defined. Whereas, viral infections often show a more severe course in obese patients, the same patients seem to have a survival benefit during bacterial infections. In particular, we summarize the main mechanical impairments in the pulmonary tract caused by obesity. Moreover, we outline the main secretory changes within the expanded adipose tissue mass, resulting in chronic low-grade inflammation. Finally, we connect these altered host factors to the influence of obesity on the development of lung infection by summarizing observations from clinical and experimental data.
Collapse
|
34
|
Abstract
The field of single nanoparticle plasmonics has grown enormously. There is no doubt that a wide diversity of the nanoplasmonic techniques and nanostructures represents a tremendous opportunity for fundamental biomedical studies as well as sensing and imaging applications. Single nanoparticle plasmonic biosensors are efficient in label-free single-molecule detection, as well as in monitoring real-time binding events of even several biomolecules. In the present review, we have discussed the prominent advantages and advances in single particle characterization and synthesis as well as new insight into and information on biomedical diagnosis uniquely obtained using single particle approaches. The approaches include the fundamental studies of nanoplasmonic behavior, two typical methods based on refractive index change and characteristic light intensity change, exciting innovations of synthetic strategies for new plasmonic nanostructures, and practical applications using single particle sensing, imaging, and tracking. The basic sphere and rod nanostructures are the focus of extensive investigations in biomedicine, while they can be programmed into algorithmic assemblies for novel plasmonic diagnosis. Design of single nanoparticles for the detection of single biomolecules will have far-reaching consequences in biomedical diagnosis.
Collapse
Affiliation(s)
- Xingyi Ma
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Korea.
| | - Sang Jun Sim
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Korea.
| |
Collapse
|
35
|
Adrover-Jaume C, Alba-Patiño A, Clemente A, Santopolo G, Vaquer A, Russell SM, Barón E, González Del Campo MDM, Ferrer JM, Berman-Riu M, García-Gasalla M, Aranda M, Borges M, de la Rica R. Paper biosensors for detecting elevated IL-6 levels in blood and respiratory samples from COVID-19 patients. SENSORS AND ACTUATORS. B, CHEMICAL 2021; 330:129333. [PMID: 33519090 PMCID: PMC7833127 DOI: 10.1016/j.snb.2020.129333] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 05/05/2023]
Abstract
Decentralizing COVID-19 care reduces contagions and affords a better use of hospital resources. We introduce biosensors aimed at detecting severe cases of COVID-19 in decentralized healthcare settings. They consist of a paper immunosensor interfaced with a smartphone. The immunosensors have been designed to generate intense colorimetric signals when the sample contains ultralow concentrations of IL-6, which has been proposed as a prognosis biomarker of COVID-19. This is achieved by combining a paper-based signal amplification mechanism with polymer-filled reservoirs for dispensing antibody-decorated nanoparticles and a bespoken app for color quantification. With this design we achieved a low limit of detection (LOD) of 10-3 pg mL-1 and semi-quantitative measurements in a wide dynamic range between 10-3 and 102 pg mL-1 in PBS. The assay time is under 10 min. The low LOD allowed us to dilute blood samples and detect IL-6 with an LOD of 1.3 pg mL-1 and a dynamic range up to 102 pg mL-1. Following this protocol, we were able to stratify COVID-19 patients according to different blood levels of IL-6. We also report on the detection of IL-6 in respiratory samples (bronchial aspirate, BAS) from COVID-19 patients. The test could be easily adapted to detect other cytokines such as TNF-α and IL-8 by changing the antibodies decorating the nanoparticles accordingly. The ability of detecting cytokines in blood and respiratory samples paves the way for monitoring local inflammation in the lungs as well as systemic inflammation levels in the body.
Collapse
Affiliation(s)
- Cristina Adrover-Jaume
- Multidisciplinary Sepsis Group, Health Research Institute of the Balearic Islands (IdISBa), Son Espases University Hospital, 07120, Palma de Mallorca, Spain
- University of the Balearic Islands, Chemistry Department, Cra. de Valldemossa km 7.5, 07122, Palma de Mallorca, Spain
| | - Alejandra Alba-Patiño
- Multidisciplinary Sepsis Group, Health Research Institute of the Balearic Islands (IdISBa), Son Espases University Hospital, 07120, Palma de Mallorca, Spain
- University of the Balearic Islands, Chemistry Department, Cra. de Valldemossa km 7.5, 07122, Palma de Mallorca, Spain
| | - Antonio Clemente
- Multidisciplinary Sepsis Group, Health Research Institute of the Balearic Islands (IdISBa), Son Espases University Hospital, 07120, Palma de Mallorca, Spain
| | - Giulia Santopolo
- Multidisciplinary Sepsis Group, Health Research Institute of the Balearic Islands (IdISBa), Son Espases University Hospital, 07120, Palma de Mallorca, Spain
- University of the Balearic Islands, Chemistry Department, Cra. de Valldemossa km 7.5, 07122, Palma de Mallorca, Spain
| | - Andreu Vaquer
- Multidisciplinary Sepsis Group, Health Research Institute of the Balearic Islands (IdISBa), Son Espases University Hospital, 07120, Palma de Mallorca, Spain
| | - Steven M Russell
- Multidisciplinary Sepsis Group, Health Research Institute of the Balearic Islands (IdISBa), Son Espases University Hospital, 07120, Palma de Mallorca, Spain
| | - Enrique Barón
- Multidisciplinary Sepsis Group, Health Research Institute of the Balearic Islands (IdISBa), Son Espases University Hospital, 07120, Palma de Mallorca, Spain
| | - María Del Mar González Del Campo
- Multidisciplinary Sepsis Group, Health Research Institute of the Balearic Islands (IdISBa), Son Espases University Hospital, 07120, Palma de Mallorca, Spain
| | - Joana M Ferrer
- Immune Response in Human Pathology Group, Health Research Institute of the Balearic Islands (IdISBa), Spain
- Immunology Department, Son Espases University Hospital, Spain Son Espases University Hospital, 07120, Palma de Mallorca, Spain
| | - María Berman-Riu
- Immune Response in Human Pathology Group, Health Research Institute of the Balearic Islands (IdISBa), Spain
| | - Mercedes García-Gasalla
- Infectious Diseases-HIV Group, Health Research Institute of the Balearic Islands (IdISBa), Son Espases University Hospital, 07120, Palma de Mallorca, Spain
| | - María Aranda
- Multidisciplinary Sepsis Group, Health Research Institute of the Balearic Islands (IdISBa), Son Espases University Hospital, 07120, Palma de Mallorca, Spain
- Multidisciplinary Sepsis Unit, ICU, Son Llàtzer University Hospital, 07198, Palma de Mallorca, Spain
| | - Marcio Borges
- Multidisciplinary Sepsis Group, Health Research Institute of the Balearic Islands (IdISBa), Son Espases University Hospital, 07120, Palma de Mallorca, Spain
- Multidisciplinary Sepsis Unit, ICU, Son Llàtzer University Hospital, 07198, Palma de Mallorca, Spain
| | - Roberto de la Rica
- Multidisciplinary Sepsis Group, Health Research Institute of the Balearic Islands (IdISBa), Son Espases University Hospital, 07120, Palma de Mallorca, Spain
- University of the Balearic Islands, Chemistry Department, Cra. de Valldemossa km 7.5, 07122, Palma de Mallorca, Spain
| |
Collapse
|
36
|
Picollet-D'hahan N, Zuchowska A, Lemeunier I, Le Gac S. Multiorgan-on-a-Chip: A Systemic Approach To Model and Decipher Inter-Organ Communication. Trends Biotechnol 2021; 39:788-810. [PMID: 33541718 DOI: 10.1016/j.tibtech.2020.11.014] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022]
Abstract
Multiorgan-on-a-chip (multi-OoC) platforms have great potential to redefine the way in which human health research is conducted. After briefly reviewing the need for comprehensive multiorgan models with a systemic dimension, we highlight scenarios in which multiorgan models are advantageous. We next overview existing multi-OoC platforms, including integrated body-on-a-chip devices and modular approaches involving interconnected organ-specific modules. We highlight how multi-OoC models can provide unique information that is not accessible using single-OoC models. Finally, we discuss remaining challenges for the realization of multi-OoC platforms and their worldwide adoption. We anticipate that multi-OoC technology will metamorphose research in biology and medicine by providing holistic and personalized models for understanding and treating multisystem diseases.
Collapse
Affiliation(s)
- Nathalie Picollet-D'hahan
- Université Grenoble Alpes, Institut National de la Santé et de la Recherche Médicale (INSERM), Commissariat à l'Energie Atomique (CEA) Interdisciplinary Research Institute of Grenoble (IRIG) Biomicrotechnology and Functional Genomics (BIOMICS), Grenoble, France.
| | - Agnieszka Zuchowska
- Applied Microfluidics for Bioengineering Research (AMBER), MESA+ Institute for Nanotechnology, TechMed Center, University of Twente, 7500AE Enschede, The Netherlands
| | - Iris Lemeunier
- Université Grenoble Alpes, Institut National de la Santé et de la Recherche Médicale (INSERM), Commissariat à l'Energie Atomique (CEA) Interdisciplinary Research Institute of Grenoble (IRIG) Biomicrotechnology and Functional Genomics (BIOMICS), Grenoble, France
| | - Séverine Le Gac
- Applied Microfluidics for Bioengineering Research (AMBER), MESA+ Institute for Nanotechnology, TechMed Center, University of Twente, 7500AE Enschede, The Netherlands.
| |
Collapse
|
37
|
Yang F, Carmona A, Stojkova K, Garcia Huitron EI, Goddi A, Bhushan A, Cohen RN, Brey EM. A 3D human adipose tissue model within a microfluidic device. LAB ON A CHIP 2021; 21:435-446. [PMID: 33351023 PMCID: PMC7876365 DOI: 10.1039/d0lc00981d] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
An accurate in vitro model of human adipose tissue could assist in the study of adipocyte function and allow for better tools for screening new therapeutic compounds. Cell culture models on two-dimensional surfaces fall short of mimicking the three-dimensional in vivo adipose environment, while three-dimensional culture models are often unable to support long-term cell culture due, in part, to insufficient mass transport. Microfluidic systems have been explored for adipose tissue models. However, current systems have primarily focused on 2D cultured adipocytes. In this work, a 3D human adipose microtissue was engineered within a microfluidic system. Human adipose-derived stem cells (ADSCs) were used as the cell source for generating differentiated adipocytes. The ADSCs differentiated within the microfluidic system formed a dense lipid-loaded mass with the expression of adipose tissue genetic markers. Engineered adipose tissue showed a decreased adiponectin secretion and increased free fatty acid secretion with increasing shear stress. Adipogenesis markers were downregulated with increasing shear stress. Overall, this microfluidic system enables the on-chip differentiation and development of a functional 3D human adipose microtissue supported by the interstitial flow. This system could potentially serve as a platform for in vitro drug testing for adipose tissue-related diseases.
Collapse
Affiliation(s)
- Feipeng Yang
- Illinois Institute of Technology, Department of Biomedical Engineering, Chicago, 60616, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Chen YT, Lee YC, Lai YH, Lim JC, Huang NT, Lin CT, Huang JJ. Review of Integrated Optical Biosensors for Point-Of-Care Applications. BIOSENSORS-BASEL 2020; 10:bios10120209. [PMID: 33353033 PMCID: PMC7766912 DOI: 10.3390/bios10120209] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/12/2020] [Accepted: 12/16/2020] [Indexed: 12/25/2022]
Abstract
This article reviews optical biosensors and their integration with microfluidic channels. The integrated biosensors have the advantages of higher accuracy and sensitivity because they can simultaneously monitor two or more parameters. They can further incorporate many functionalities such as electrical control and signal readout monolithically in a single semiconductor chip, making them ideal candidates for point-of-care testing. In this article, we discuss the applications by specifically looking into point-of-care testing (POCT) using integrated optical sensors. The requirement and future perspective of integrated optical biosensors for POC is addressed.
Collapse
Affiliation(s)
- Yung-Tsan Chen
- Graduate Institute of Photonics and Optoelectronics, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan; (Y.-T.C.); (Y.-C.L.); (Y.-H.L.); (J.-C.L.)
| | - Ya-Chu Lee
- Graduate Institute of Photonics and Optoelectronics, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan; (Y.-T.C.); (Y.-C.L.); (Y.-H.L.); (J.-C.L.)
| | - Yao-Hsuan Lai
- Graduate Institute of Photonics and Optoelectronics, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan; (Y.-T.C.); (Y.-C.L.); (Y.-H.L.); (J.-C.L.)
| | - Jin-Chun Lim
- Graduate Institute of Photonics and Optoelectronics, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan; (Y.-T.C.); (Y.-C.L.); (Y.-H.L.); (J.-C.L.)
| | - Nien-Tsu Huang
- Department of Electrical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan; (N.-T.H.); (C.-T.L.)
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan
| | - Chih-Ting Lin
- Department of Electrical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan; (N.-T.H.); (C.-T.L.)
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan
- Graduate Institute of Electronics Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan
| | - Jian-Jang Huang
- Graduate Institute of Photonics and Optoelectronics, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan; (Y.-T.C.); (Y.-C.L.); (Y.-H.L.); (J.-C.L.)
- Department of Electrical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan; (N.-T.H.); (C.-T.L.)
- Correspondence:
| |
Collapse
|
39
|
Ma C, Peng Y, Li H, Chen W. Organ-on-a-Chip: A New Paradigm for Drug Development. Trends Pharmacol Sci 2020; 42:119-133. [PMID: 33341248 DOI: 10.1016/j.tips.2020.11.009] [Citation(s) in RCA: 241] [Impact Index Per Article: 48.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 01/16/2023]
Abstract
The pharmaceutical industry has been desperately searching for efficient drug discovery methods. Organ-on-a-Chip, a cutting-edge technology that can emulate the physiological environment and functionality of human organs on a chip for disease modeling and drug testing, shows great potential for revolutionizing the drug development pipeline. However, successful translation of this novel engineering platform into routine pharmacological and medical scenarios remains to be realized. In this review, we discuss how the Organ-on-a-Chip technology can have critical roles in different preclinical stages of drug development and highlight the current challenges in translation and commercialization of this technology for the pharmacological and medical end-users. Moreover, this review sheds light on the future developmental trends and need for a next-generation Organ-on-a-Chip platform to bridge the gap between animal studies and clinical trials for the pharmaceutical industry.
Collapse
Affiliation(s)
- Chao Ma
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY 11201, USA; Department of Biomedical Engineering, New York University, Brooklyn, NY 11201, USA
| | - Yansong Peng
- Department of Biomedical Engineering, New York University, Brooklyn, NY 11201, USA
| | - Hongtong Li
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY 11201, USA
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY 11201, USA; Department of Biomedical Engineering, New York University, Brooklyn, NY 11201, USA; Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA.
| |
Collapse
|
40
|
Pope BD, Warren CR, Dahl MO, Pizza CV, Henze DE, Sinatra NR, Gonzalez GM, Chang H, Liu Q, Glieberman AL, Ferrier JP, Cowan CA, Parker KK. Fattening chips: hypertrophy, feeding, and fasting of human white adipocytes in vitro. LAB ON A CHIP 2020; 20:4152-4165. [PMID: 33034335 PMCID: PMC7818847 DOI: 10.1039/d0lc00508h] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Adipose is a distributed organ that performs vital endocrine and energy homeostatic functions. Hypertrophy of white adipocytes is a primary mode of both adaptive and maladaptive weight gain in animals and predicts metabolic syndrome independent of obesity. Due to the failure of conventional culture to recapitulate adipocyte hypertrophy, technology for production of adult-size adipocytes would enable applications such as in vitro testing of weight loss therapeutics. To model adaptive adipocyte hypertrophy in vitro, we designed and built fat-on-a-chip using fiber networks inspired by extracellular matrix in adipose tissue. Fiber networks extended the lifespan of differentiated adipocytes, enabling growth to adult sizes. By micropatterning preadipocytes in a native cytoarchitecture and by adjusting cell-to-cell spacing, rates of hypertrophy were controlled independent of culture time or differentiation efficiency. In vitro hypertrophy followed a nonlinear, nonexponential growth model similar to human development and elicited transcriptomic changes that increased overall similarity with primary tissue. Cells on the chip responded to simulated meals and starvation, which potentiated some adipocyte endocrine and metabolic functions. To test the utility of the platform for therapeutic development, transcriptional network analysis was performed, and retinoic acid receptors were identified as candidate drug targets. Regulation by retinoid signaling was suggested further by pharmacological modulation, where activation accelerated and inhibition slowed hypertrophy. Altogether, this work presents technology for mature adipocyte engineering, addresses the regulation of cell growth, and informs broader applications for synthetic adipose in pharmaceutical development, regenerative medicine, and cellular agriculture.
Collapse
Affiliation(s)
- Benjamin D Pope
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA. and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Curtis R Warren
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Madeleine O Dahl
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - Christina V Pizza
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - Douglas E Henze
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - Nina R Sinatra
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - Grant M Gonzalez
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - Huibin Chang
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - Qihan Liu
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - Aaron L Glieberman
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - John P Ferrier
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - Chad A Cowan
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA and Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Kevin Kit Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA. and Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| |
Collapse
|
41
|
Maschmeyer I, Kakava S. Organ-on-a-Chip. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2020; 179:311-342. [PMID: 32948885 DOI: 10.1007/10_2020_135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Limitations of the current tools used in the drug development process, cell cultures, and animal models have highlighted the need for a new powerful tool that can emulate the human physiology in vitro. Advances in the field of microfluidics have made the realization of this tool closer than ever. Organ-on-a-chip platforms have been the first step forward, leading to the combination and integration of multiple organ models in the same platform with human-on-a-chip being the ultimate goal. Despite the current progress and technological developments, there are still several unmet engineering and biological challenges curtailing their development and widespread application in the biomedical field. The potentials, challenges, and current work on this unprecedented tool are being discussed in this chapter.
Collapse
|
42
|
Vohra MS, Ahmad B, Serpell CJ, Parhar IS, Wong EH. Murine in vitro cellular models to better understand adipogenesis and its potential applications. Differentiation 2020; 115:62-84. [PMID: 32891960 DOI: 10.1016/j.diff.2020.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/08/2020] [Accepted: 08/13/2020] [Indexed: 02/07/2023]
Abstract
Adipogenesis has been extensively studied using in vitro models of cellular differentiation, enabling long-term regulation of fat cell metabolism in human adipose tissue (AT) material. Many studies promote the idea that manipulation of this process could potentially reduce the prevalence of obesity and its related diseases. It has now become essential to understand the molecular basis of fat cell development to tackle this pandemic disease, by identifying therapeutic targets and new biomarkers. This review explores murine cell models and their applications for study of the adipogenic differentiation process in vitro. We focus on the benefits and limitations of different cell line models to aid in interpreting data and selecting a good cell line model for successful understanding of adipose biology.
Collapse
Affiliation(s)
- Muhammad Sufyan Vohra
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia.
| | - Bilal Ahmad
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia.
| | - Christopher J Serpell
- School of Physical Sciences, Ingram Building, University of Kent, Canterbury, Kent, CT2 7NH, United Kingdom.
| | - Ishwar S Parhar
- Brain Research Institute, Jeffery Cheah School of Medicine and Health Sciences, Monash University, Bandar Sunway, PJ 47500, Selangor, Malaysia.
| | - Eng Hwa Wong
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
43
|
Ferhan AR, Yoon BK, Jeon WY, Cho NJ. Biologically interfaced nanoplasmonic sensors. NANOSCALE ADVANCES 2020; 2:3103-3114. [PMID: 36134263 PMCID: PMC9418064 DOI: 10.1039/d0na00279h] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 06/26/2020] [Indexed: 05/30/2023]
Abstract
Understanding biointerfacial processes is crucial in various fields across fundamental and applied biology, but performing quantitative studies via conventional characterization techniques remains challenging due to instrumentation as well as analytical complexities and limitations. In order to accelerate translational research and address current challenges in healthcare and medicine, there is an outstanding need to develop surface-sensitive technologies with advanced measurement capabilities. Along this line, nanoplasmonic sensing has emerged as a powerful tool to quantitatively study biointerfacial processes owing to its high spatial resolution at the nanoscale. Consequently, the development of robust biological interfacing strategies becomes imperative to maximize its characterization potential. This review will highlight and discuss the critical role of biological interfacing within the context of constructing nanoplasmonic sensing platforms for biointerfacial science applications. Apart from paving the way for the development of highly surface-sensitive characterization tools that will spur fundamental biological interaction studies and improve the overall understanding of biological processes, the basic principles behind biointerfacing strategies presented in this review are also applicable to other fields that involve an interface between an inorganic material and a biological system.
Collapse
Affiliation(s)
- Abdul Rahim Ferhan
- School of Materials Science and Engineering, Nanyang Technological University 50 Nanyang Avenue 639798 Singapore
| | - Bo Kyeong Yoon
- School of Materials Science and Engineering, Nanyang Technological University 50 Nanyang Avenue 639798 Singapore
- School of Chemical Engineering, Sungkyunkwan University Suwon 16419 Republic of Korea
| | - Won-Yong Jeon
- School of Chemical Engineering, Sungkyunkwan University Suwon 16419 Republic of Korea
| | - Nam-Joon Cho
- School of Materials Science and Engineering, Nanyang Technological University 50 Nanyang Avenue 639798 Singapore
| |
Collapse
|
44
|
Ramadan Q, Zourob M. Organ-on-a-chip engineering: Toward bridging the gap between lab and industry. BIOMICROFLUIDICS 2020; 14:041501. [PMID: 32699563 PMCID: PMC7367691 DOI: 10.1063/5.0011583] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/22/2020] [Indexed: 05/03/2023]
Abstract
Organ-on-a-chip (OOC) is a very ambitious emerging technology with a high potential to revolutionize many medical and industrial sectors, particularly in preclinical-to-clinical translation in the pharmaceutical arena. In vivo, the function of the organ(s) is orchestrated by a complex cellular structure and physiochemical factors within the extracellular matrix and secreted by various types of cells. The trend in in vitro modeling is to simplify the complex anatomy of the human organ(s) to the minimal essential cellular structure "micro-anatomy" instead of recapitulating the full cellular milieu that enables studying the absorption, metabolism, as well as the mechanistic investigation of drug compounds in a "systemic manner." However, in order to reflect the human physiology in vitro and hence to be able to bridge the gap between the in vivo and in vitro data, simplification should not compromise the physiological relevance. Engineering principles have long been applied to solve medical challenges, and at this stage of organ-on-a-chip technology development, the work of biomedical engineers, focusing on device engineering, is more important than ever to accelerate the technology transfer from the academic lab bench to specialized product development institutions and to the increasingly demanding market. In this paper, instead of presenting a narrative review of the literature, we systemically present a synthesis of the best available organ-on-a-chip technology from what is found, what has been achieved, and what yet needs to be done. We emphasized mainly on the requirements of a "good in vitro model that meets the industrial need" in terms of the structure (micro-anatomy), functions (micro-physiology), and characteristics of the device that hosts the biological model. Finally, we discuss the biological model-device integration supported by an example and the major challenges that delay the OOC technology transfer to the industry and recommended possible options to realize a functional organ-on-a-chip system.
Collapse
Affiliation(s)
- Qasem Ramadan
- Alfaisal University, Al Zahrawi Street, Riyadh 11533, Kingdom of Saudi Arabia
| | - Mohammed Zourob
- Alfaisal University, Al Zahrawi Street, Riyadh 11533, Kingdom of Saudi Arabia
| |
Collapse
|
45
|
Hu J, Li X, Judd RL, Easley CJ. Rapid lipolytic oscillations in ex vivo adipose tissue explants revealed through microfluidic droplet sampling at high temporal resolution. LAB ON A CHIP 2020; 20:1503-1512. [PMID: 32239045 PMCID: PMC7380261 DOI: 10.1039/d0lc00103a] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Our understanding of adipose tissue biology has steadily evolved. While structural and energy storage functionalities have been in the forefront, a key endocrine role for adipocytes was revealed only over the last few decades. In contrast to the wealth of information on dynamic function of other endocrine tissues, few studies have focused on dynamic adipose tissue function or on tool development toward that end. Here, we apply our unique droplet-based microfluidic devices to culture, perfuse, and sample secretions from primary murine epididymal white adipose tissue (eWAT), and from predifferentiated clusters of 3T3-L1 adipocytes. Through automated control, oil-segmented aqueous droplets (∼2.6 nL) were sampled from tissue or cells at 3.5 second temporal resolution (including sample and reference droplets), with integrated enzyme assays enabling real-time quantification of glycerol (down to 1.9 fmol per droplet). This high resolution revealed previously unreported oscillations in secreted glycerol at frequencies of 0.2 to 2.0 min-1 (∼30-300 s periods) present in the primary tissue but not in clustered cells. Low-level bursts (∼50 fmol) released in basal conditions were contrasted with larger bursts (∼300 fmol) during stimulation. Further, both fold changes and burst magnitudes were decreased in eWAT of aged and obese mice. These results, combined with immunostaining and photobleaching analyses, suggest that gap-junctional coupling or nerve cell innervation within the intact ex vivo tissue explants play important roles in this apparent tissue-level, lipolytic synchronization. High-resolution, quantitative sampling by droplet microfluidics thus permitted unique biological information to be observed, giving an analytical framework poised for future studies of dynamic oscillatory function of adipose and other tissues.
Collapse
Affiliation(s)
- Juan Hu
- Department of Chemistry and Biochemistry, Auburn University, Auburn, AL 36849, USA.
| | | | | | | |
Collapse
|
46
|
Rodriguez-Moncayo R, Jimenez-Valdes RJ, Gonzalez-Suarez AM, Garcia-Cordero JL. Integrated Microfluidic Device for Functional Secretory Immunophenotyping of Immune Cells. ACS Sens 2020; 5:353-361. [PMID: 31927915 DOI: 10.1021/acssensors.9b01786] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Integrated platforms for automatic assessment of cellular functional secretory immunophenotyping could have a widespread use in the diagnosis, real-time monitoring, and therapy evaluation of several pathologies. We present a microfluidic platform with integrated biosensors and culture chambers to measure cytokine secretion from a consistent and uniform number of immune cells. The biosensor relies on a fluorescence sandwich immunoassay enabled by the mechanically induced trapping of molecular interactions method. The platform contains 32 cell culture chambers, each patterned with an array of 492 microwells, to capture and analyze both adherent and nonadherent immune cells. Multiple stimuli can be delivered to a set of culture chambers. Per chamber, we were able to capture consistently 1113 ± 191 of blood-derived monocytes and neutrophils and 348 ± 37 THP-1 monocytes. Good occupancy efficiencies of ∼70% with a uniformity of ∼90% across all of the culture chambers of the device were achieved. Furthermore, we demonstrate that up to 96% of cells remain viable for the first 48 h. The employment of epoxy-modified glass substrates and active mixing enhanced the biosensing performance compared to the use of bare glass and simple diffusion. Finally, we performed functional secretory analysis of interleukin-8 and tumor necrosis factor alpha from human neutrophils and monocytes, stimulated with various doses of lipopolysaccharide and phorbol 12-myristate 13-acetate-ionomycin, respectively. We foresee the employment of our microfluidic platform in the diagnosis of different pathologies where alterations in cytokine secretion patterns can be used as biomarkers.
Collapse
Affiliation(s)
- Roberto Rodriguez-Moncayo
- Unidad Monterrey, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Parque PIIT, Apodaca, Nuevo León 66628, Mexico
| | - Rocio Jimena Jimenez-Valdes
- Unidad Monterrey, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Parque PIIT, Apodaca, Nuevo León 66628, Mexico
| | - Alan Mauricio Gonzalez-Suarez
- Unidad Monterrey, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Parque PIIT, Apodaca, Nuevo León 66628, Mexico
| | - Jose Luis Garcia-Cordero
- Unidad Monterrey, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Parque PIIT, Apodaca, Nuevo León 66628, Mexico
| |
Collapse
|
47
|
Wang C, Cai Y, MacLACHLAN A, Chen P. Novel Nanoplasmonic-Structure-Based Integrated Microfluidic Biosensors for Label-Free in Situ Immune Functional Analysis: A review of recent progress. IEEE NANOTECHNOLOGY MAGAZINE 2020; 14:46-C3. [PMID: 34290843 DOI: 10.1109/mnano.2020.2966205] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Chuanyu Wang
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Alabama
| | - Yuxin Cai
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Alabama
| | - Alana MacLACHLAN
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Alabama
| | - Pengyu Chen
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Alabama
| |
Collapse
|
48
|
Lee SN, Choi JH, Cho HY, Choi JW. Metallic Nanoparticle-Based Optical Cell Chip for Nondestructive Monitoring of Intra/Extracellular Signals. Pharmaceutics 2020; 12:pharmaceutics12010050. [PMID: 31936079 PMCID: PMC7022866 DOI: 10.3390/pharmaceutics12010050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 12/31/2019] [Accepted: 01/06/2020] [Indexed: 12/23/2022] Open
Abstract
The biosensing platform is noteworthy for high sensitivity and precise detection of target analytes, which are related to the status of cells or specific diseases. The modification of the transducers with metallic nanoparticles (MNPs) has attracted attention owing to excellent features such as improved sensitivity and selectivity. Moreover, the incorporation of MNPs into biosensing systems may increase the speed and the capability of the biosensors. In this review, we introduce the current progress of the developed cell-based biosensors, cell chip, based on the unique physiochemical features of MNPs. Mainly, we focus on optical intra/extracellular biosensing methods, including fluorescence, localized surface plasmon resonance (LSPR), and surface-enhanced Raman spectroscopy (SERS) based on the coupling of MNPs. We believe that the topics discussed here are useful and able to provide a guideline in the development of new MNP-based cell chip platforms for pharmaceutical applications such as drug screening and toxicological tests in the near future.
Collapse
Affiliation(s)
- Sang-Nam Lee
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-Gu, Seoul 04107, Korea; (S.-N.L.); (J.-H.C.)
- Uniance Gene Inc., 1107 Teilhard Hall, 35 Baekbeom-Ro, Mapo-Gu, Seoul 04107, Korea
| | - Jin-Ha Choi
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-Gu, Seoul 04107, Korea; (S.-N.L.); (J.-H.C.)
| | - Hyeon-Yeol Cho
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-Gu, Seoul 04107, Korea; (S.-N.L.); (J.-H.C.)
- Correspondence: (H.-Y.C.); (J.-W.C.); Tel.: +82-2-705-8480 (J.-W.C.)
| | - Jeong-Woo Choi
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-Gu, Seoul 04107, Korea; (S.-N.L.); (J.-H.C.)
- Correspondence: (H.-Y.C.); (J.-W.C.); Tel.: +82-2-705-8480 (J.-W.C.)
| |
Collapse
|
49
|
Sukumar P, Deliorman M, Brimmo AT, Alnemari R, Elsori D, Chen W, Qasaimeh MA. Airplug-mediated isolation and centralization of single T cells in rectangular microwells for biosensing. ADVANCED THERAPEUTICS 2020; 3:1900085. [PMID: 33117882 PMCID: PMC7591138 DOI: 10.1002/adtp.201900085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Indexed: 11/09/2022]
Abstract
Sorting cells in a single cell per microwell format is of great interest to basic biology studies, biotherapeutics, and biosensing including cell phenotyping. For instance, isolation of individual immune T cells in rectangular microwells has been shown to empower the multiplex cytokine profiling at the single cell level for therapeutics applications. The present study, however, shows that there is an existing bias in temporal cytokine sensing that originates from random "unpredicted" positions of loaded cells within the rectangular microwells. To eliminate this bias, the isolated cells need to be well-aligned with each other and relative to the sensing elements. Hence, an approach that utilizes the in situ formation and release of airplugs to localize cells towards the center of the rectangular microwells is reported. The chip includes 2250 microwells (each 500 × 50 × 20 μm3) arranged in 9 rows. Results showed 20% efficiency in trapping single T cells per microwells, where cells are localized within ±3% of the center of microwells. The developed platform could provide real-time dynamic and unbiased multiplex cytokine detection from single T cells for phenotyping and biotherapeutics studies.
Collapse
Affiliation(s)
- Pavithra Sukumar
- Division of Engineering, New York University Abu Dhabi (NYUAD), P.O. Box 129188, Abu Dhabi, UAE
| | - Muhammedin Deliorman
- Division of Engineering, New York University Abu Dhabi (NYUAD), P.O. Box 129188, Abu Dhabi, UAE
| | - Ayoola T Brimmo
- Division of Engineering, New York University Abu Dhabi (NYUAD), P.O. Box 129188, Abu Dhabi, UAE
| | - Roaa Alnemari
- Division of Engineering, New York University Abu Dhabi (NYUAD), P.O. Box 129188, Abu Dhabi, UAE
| | - Deena Elsori
- Department of Applied Sciences and Mathematics, Abu Dhabi University, P.O. Box 59911, Abu Dhabi, UAE
| | - Weiqiang Chen
- Department of Biomedical Engineering, New York University, Brooklyn, NY 11201, USA
| | - Mohammad A Qasaimeh
- Division of Engineering, New York University Abu Dhabi (NYUAD), P.O. Box 129188, Abu Dhabi, UAE
| |
Collapse
|
50
|
Yesilkoy F. Optical Interrogation Techniques for Nanophotonic Biochemical Sensors. SENSORS 2019; 19:s19194287. [PMID: 31623315 PMCID: PMC6806184 DOI: 10.3390/s19194287] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/25/2019] [Accepted: 09/27/2019] [Indexed: 12/14/2022]
Abstract
The manipulation of light via nanoengineered surfaces has excited the optical community in the past few decades. Among the many applications enabled by nanophotonic devices, sensing has stood out due to their capability of identifying miniscule refractive index changes. In particular, when free-space propagating light effectively couples into subwavelength volumes created by nanostructures, the strongly-localized near-fields can enhance light’s interaction with matter at the nanoscale. As a result, nanophotonic sensors can non-destructively detect chemical species in real-time without the need of exogenous labels. The impact of such nanophotonic devices on biochemical sensor development became evident as the ever-growing research efforts in the field started addressing many critical needs in biomedical sciences, such as low-cost analytical platforms, simple quantitative bioassays, time-resolved sensing, rapid and multiplexed detection, single-molecule analytics, among others. In this review, the optical transduction methods used to interrogate optical resonances of nanophotonic sensors will be highlighted. Specifically, the optical methodologies used thus far will be evaluated based on their capability of addressing key requirements of the future sensor technologies, including miniaturization, multiplexing, spatial and temporal resolution, cost and sensitivity.
Collapse
Affiliation(s)
- Filiz Yesilkoy
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|