1
|
Zhao C, Zhang Z, Li J, Lu Y, Ma F, Wang Z, Geng J, Huang B, Qin Y. Development of a Quick and Highly Sensitive Amplified Luminescent Proximity Homogeneous Assay for Detection of Saxitoxin in Shellfish. Toxins (Basel) 2024; 16:341. [PMID: 39195751 PMCID: PMC11360761 DOI: 10.3390/toxins16080341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 08/29/2024] Open
Abstract
Saxitoxin (STX), an exceptionally potent marine toxin for which no antidote is currently available, is produced by methanogens and cyanobacteria. This poses a significant threat to both shellfish aquaculture and human health. Consequently, the development of a rapid, highly sensitive STX detection method is of great significance. The objective of this research is to create a novel approach for identifying STX. Therefore, amplified luminescent proximity homogeneous assay (AlphaLISA) was established using a direct competition method based on the principles of fluorescence resonance energy transfer and antigen-antibody specific binding. This method is sensitive, rapid, performed without washing, easy to operate, and can detect 8-128 ng/mL of STX in only 10 min. The limit of detection achieved by this method is as low as 4.29 ng/mL with coefficients of variation for the intra-batch and inter-batch analyses ranging from 2.61% to 3.63% and from 7.67% to 8.30%, respectively. In conclusion, our study successfully establishes a simple yet sensitive, rapid, and accurate AlphaLISA method for the detection of STX which holds great potential in advancing research on marine biotoxins.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Biao Huang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (C.Z.); (Z.Z.); (J.L.); (Y.L.); (Z.W.); (J.G.)
| | - Yuan Qin
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (C.Z.); (Z.Z.); (J.L.); (Y.L.); (Z.W.); (J.G.)
| |
Collapse
|
2
|
Zong H, Shang X, Wang X, Chen T, Wang Y, Ren Y, Jiang Y, Li Y, Lv Q, Liu P. Diagnosis of septic shock by serum measurement of human neutrophil lipocalin by a rapid homogeneous assay. J Immunol Methods 2023; 522:113570. [PMID: 37774777 DOI: 10.1016/j.jim.2023.113570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 10/01/2023]
Abstract
BACKGROUND Human neutrophil lipocalin (HNL) is a marker of neutrophil activation and has a high efficacy in diagnosing bacterial infections. In this study, we applied the AlphaLISA technique to measure the serum level of HNL, evaluate HNL's efficacy in diagnosing septic shock, and identify any association between HNL level and septic patients' prognosis. METHODS We collected 146 serum samples from the Fifth Medical Center of Chinese PLA General Hospital. HNL was measured by AlphaLISA and results were compared with commercial ELISA kits. We studied 78 patients admitted to the ICU with sepsis and data on their clinical and physiological characteristics were recorded. Blood levels of HNL, procalcitonin (PCT), high-sensitivity C-reactive protein (hs-CRP), and lactate were measured. A receiver operating characteristic (ROC) curve was used to evaluate the performance of each marker. RESULTS The AlphaLISA assay for serum HNL had a detection range from 1.5 ng/mL to 1000 ng/mL, with a detection limit of 1 ng/mL and a detection time of approximately 25 min. The AlphaLISA assay's results were in high agreement with ELISA results (R2 = 0.9413). HNL levels were analyzed in sepsis patients, and HNL was significantly higher in sepsis patients with shock compared to sepsis patients without shock (median 356.47 ng/mL vs 158.93 ng/mL, P < 0.0001) and in the 28-day non-survivor group compared to the 28-day survivor group (median 331.83 ng/mL vs 175.17 ng/mL, P < 0.0001). ROC curve analysis was performed for the biomarkers. In differentiating the diagnosis of septic shock from sepsis patients, HNL was the most effective marker (AUC = 0.857), followed by PCT (AUC = 0.754) and hs-CRP (AUC = 0.627). In predicting the prognosis of septic patients, lactate had the best effect (AUC = 0.805), followed by HNL (AUC = 0.784), PCT (AUC = 0.721), and hs-CRP (AUC = 0.583). CONCLUSIONS As an assessment tool, we found that our AlphaLISA had good consistency with an ELISA and had several other advantages, including requiring a shorter processing time and detecting a wider range of serum HNL concentrations. Monitoring serum HNL levels of patients admitted to the ICU might be useful in distinguishing sepsis patients who have septic shock from other sepsis patients, indicating its value in the prediction of sepsis patient prognosis.
Collapse
Affiliation(s)
- Huijun Zong
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; The Fifth School of Clinical Medicine, Anhui Medical University, Hefei 230032, China; Department of Critical Care Medicine, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Xueyi Shang
- Department of Critical Care Medicine, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Xin Wang
- Department of Critical Care Medicine, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Ting Chen
- The Fifth School of Clinical Medicine, Anhui Medical University, Hefei 230032, China; Department of Critical Care Medicine, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Ye Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China
| | - Yuhao Ren
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China
| | - Yongqiang Jiang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China
| | - Yan Li
- The Fifth School of Clinical Medicine, Anhui Medical University, Hefei 230032, China; Department of Critical Care Medicine, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China.
| | - Qingyu Lv
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China.
| | - Peng Liu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China.
| |
Collapse
|
3
|
Li Z, Zhai W, Wang L, Liu J, Li C, Xu L. Preparation and characterization of a homogeneous immunoassay for point-of-care testing (POCT) of procalcitonin (PCT). ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:5002-5009. [PMID: 37728429 DOI: 10.1039/d3ay00890h] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Abstract
Procalcitonin (PCT) has been recognized as a specific and early marker for microbial infection and sepsis. Sensitive measuring interaction-triggered luminescence experiment (SMILE), a homogeneous immunoassay method, was established for point-of-care testing (POCT) of PCT. SMILE is achieved through the principle of double antibody sandwich, where two antibodies immobilized on the surface of polystyrene microspheres (donor and acceptor beads) bind to the PCT antigen. The donor bead contains phthalocyanine dye (luminol chemiluminescent substance) and the acceptor bead contains dimethylthiophene derivatives and Eu chelates. Therefore, singlet oxygen can be transferred when the distance between donor and acceptor beads is within 200 nm, generating detectable luminescent signals. Scanning electron microscopy (SEM) was used to detect the diameter and polymer dispersity index (PDI) of microspheres before and after binding with antibodies to characterize the immobilization of antibodies. The reaction conditions for antibody immobilization including pH, mass ratio and reaction time have also been optimized. The limit of quantitation (LOQ) of the SMILE method (0.01 ng mL-1) was lower than that of the LFI method (0.1 ng mL-1), the working range (0.01-500 ng mL-1) was wider than that of the LFI method (0.1-50 ng mL-1), and the assay time (10 min) was shorter than that of the LFI method (15 min). So, SMILE is more suitable for POCT of PCT compared with lateral flow immunochromatography (LFI), which is the most used measuring method, due to its advantages of simple operation, saving time, convenience, wide detection range, and high sensitivity and accuracy.
Collapse
Affiliation(s)
- Zhaoying Li
- Graduate School, Tianjin Medical University, Tianjin, 300070, China.
- Tianjin Chest Hospital, Tianjin, 300222, China
| | | | - Lu Wang
- Graduate School, Tianjin Medical University, Tianjin, 300070, China.
- School of Pharmacy, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Jiyang Liu
- Epsilon Biotechnology Corporation, Zhejiang, 311199, China
| | - Chunjie Li
- Tianjin Chest Hospital, Tianjin, 300222, China
| | - Liang Xu
- Graduate School, Tianjin Medical University, Tianjin, 300070, China.
- Tianjin Medical College, Tianjin, 300222, China
- School of Pharmacy, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China
| |
Collapse
|
4
|
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
|
5
|
Kneidl AM, Marth CD, Kirsch S, Weber F, Zablotski Y, Helfrich AL, Schabmeyer ST, Schneider JK, Petzl W, Zerbe H, Meyerholz-Wohllebe MM. Is the IL1RA/IL1B Ratio a Suitable Biomarker for Subclinical Endometritis in Dairy Cows? Animals (Basel) 2022; 12:ani12233363. [PMID: 36496884 PMCID: PMC9737067 DOI: 10.3390/ani12233363] [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: 10/14/2022] [Revised: 11/11/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
The adequate expression of cytokines is essential for the prevention and healing of bovine endometrial inflammation. This study investigated the intra-uterine concentration of the proinflammatory cytokine interleukin (IL)1B and its antagonist IL1RA in cows with and without subclinical endometritis (SE). Samples were taken from 37 uteri at the abattoir and 26 uteri in vivo. Uterine secretion samples were classified as showing no signs of SE (SEneg; polymorphonuclear neutrophil granulocyte (PMN) < 5%) or showing signs of SE (SEpos; PMN ≥ 5%). Concentrations and ratios for IL1B and IL1RA were measured using a commercial and a newly established AlphaLISA kit, respectively. In both groups, a higher concentration of IL1B was detected in the SEpos group compared with the SEneg group (abattoir: p = 0.027; in vivo p < 0.001). No significant differences were observed in the concentration of IL1RA (p > 0.05). In uterine secretion samples retrieved in vivo, a lower IL1RA/IL1B ratio was detected in the SEpos group compared with the SEneg group (p = 0.002). The results of this study highlight the important role of IL1B and IL1RA during endometritis and the potential of the IL1RA/IL1B ratio as a possible biomarker for SE.
Collapse
Affiliation(s)
- Anna Maria Kneidl
- Clinic for Ruminants with Ambulatory and Herd Health Services, Centre for Clinical Veterinary Medicine, Ludwig Maximilian University Munich, 85764 Oberschleissheim, Germany
| | - Christina Deborah Marth
- Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Werribee, VIC 3030, Australia
- Correspondence: ; Tel.: +61-3-8001-2641
| | - Sandra Kirsch
- Clinic for Ruminants with Ambulatory and Herd Health Services, Centre for Clinical Veterinary Medicine, Ludwig Maximilian University Munich, 85764 Oberschleissheim, Germany
| | - Frank Weber
- Clinic for Ruminants with Ambulatory and Herd Health Services, Centre for Clinical Veterinary Medicine, Ludwig Maximilian University Munich, 85764 Oberschleissheim, Germany
| | - Yury Zablotski
- Clinic for Ruminants with Ambulatory and Herd Health Services, Centre for Clinical Veterinary Medicine, Ludwig Maximilian University Munich, 85764 Oberschleissheim, Germany
| | - Anika Luzia Helfrich
- Clinic for Ruminants with Ambulatory and Herd Health Services, Centre for Clinical Veterinary Medicine, Ludwig Maximilian University Munich, 85764 Oberschleissheim, Germany
| | - Simone Tamara Schabmeyer
- Clinic for Ruminants with Ambulatory and Herd Health Services, Centre for Clinical Veterinary Medicine, Ludwig Maximilian University Munich, 85764 Oberschleissheim, Germany
| | - Julia Katharina Schneider
- Clinic for Ruminants with Ambulatory and Herd Health Services, Centre for Clinical Veterinary Medicine, Ludwig Maximilian University Munich, 85764 Oberschleissheim, Germany
| | - Wolfram Petzl
- Clinic for Ruminants with Ambulatory and Herd Health Services, Centre for Clinical Veterinary Medicine, Ludwig Maximilian University Munich, 85764 Oberschleissheim, Germany
| | - Holm Zerbe
- Clinic for Ruminants with Ambulatory and Herd Health Services, Centre for Clinical Veterinary Medicine, Ludwig Maximilian University Munich, 85764 Oberschleissheim, Germany
| | - Marie Margarete Meyerholz-Wohllebe
- Clinic for Ruminants with Ambulatory and Herd Health Services, Centre for Clinical Veterinary Medicine, Ludwig Maximilian University Munich, 85764 Oberschleissheim, Germany
| |
Collapse
|
6
|
Kim SK, Oh YH, Ko DH, Sung H, Oh HB, Hwang SH. Nanoparticle-Based Visual Detection of Amplified DNA for Diagnosis of Hepatitis C Virus. BIOSENSORS 2022; 12:bios12090744. [PMID: 36140129 PMCID: PMC9496050 DOI: 10.3390/bios12090744] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/03/2022] [Accepted: 09/08/2022] [Indexed: 12/15/2022]
Abstract
Rapid, simple, and inexpensive diagnostic point-of-care tests (POCTs) are essential for controlling infectious diseases in resource-limited settings. In this study, we developed a new detection system based on nanoparticle–DNA aggregation (STat aggregation of tagged DNA, STAT-DNA) to yield a visual change that can be easily detected by the naked eye. This simplified optical detection system was applied to detect hepatitis C virus (HCV). Reverse transcription-polymerase chain reaction (RT-PCR) was performed using primers labeled with biotin and digoxigenin. Streptavidin-coated magnetic particles (1 μm) and anti-digoxigenin antibody-coated polystyrene particles (250–350 nm) were added to form aggregates. The limit of detection (LoD) and analytical specificity were analyzed. The STAT-DNA results were compared with those of the standard real-time PCR assay using serum samples from 54 patients with hepatitis C. We achieved visualization of amplified DNA with the naked eye by adding nanoparticles to the PCR mixture without employing centrifugal force, probe addition, incubation, or dilution. The LoD of STAT-DNA was at least 101 IU/mL. STAT-DNA did not show cross-reactivity with eight viral pathogens. The detection using STAT-DNA was consistent with that using standard real-time PCR.
Collapse
Affiliation(s)
- Soo-Kyung Kim
- Department of Laboratory Medicine, Ewha Womans University College of Medicine, Seoul 07985, Korea
| | - Yoon-Hee Oh
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Dae-Hyun Ko
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Heungsup Sung
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Heung-Bum Oh
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Sang-Hyun Hwang
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Correspondence: ; Tel.: +82-2-3010-4502
| |
Collapse
|
7
|
Wang Y, Zheng Y, Li Y, Zhang S, Wang X, Zong H, Huang W, Kong D, Jiang Y, Liu P, Lv Q, Jiang H. Development of a rapid homogeneous immunoassay for detection of rotavirus in stool samples. Front Public Health 2022; 10:975720. [PMID: 35991049 PMCID: PMC9386352 DOI: 10.3389/fpubh.2022.975720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 07/19/2022] [Indexed: 11/16/2022] Open
Abstract
Rotavirus is the main pathogen causing acute viral gastroenteritis. Accurate and rapid diagnosis of rotavirus infection is important to determine appropriate treatment, prevention of unnecessary antibiotics use and control of infection spread. In this study, we established a rapid, accurate, and sensitive amplified luminescent proximity homogeneous assay linked immunosorbent assay (AlphaLISA) for detecting rotavirus and evaluated its efficacy in human stool samples. Our results demonstrated that the sensitivity of AlphaLISA (5−8) significantly exceeded that of the immunochromatographic assay (ICA, 5−4) for rotavirus antigen detection. The intra-assay and inter-assay coefficients of variation were 2.99–3.85% and 5.27–6.51%, respectively. Furthermore, AlphaLISA was specific for rotavirus and did not cross-react with other common diarrhea viruses. AlphaLISA and real-time reverse transcription polymerase chain reaction (RT-qPCR, which is considered a gold standard for detecting diarrhea viruses) tests showed consistent results on 235 stool samples, with an overall consistency rate of 97.87% and a kappa value of 0.894 (P < 0.001). The overall consistency rate of ICA compared with RT-qPCR was 95.74%. AlphaLISA showed better consistency with RT-qPCR than the routinely used ICA for rotavirus detection in stool samples. The AlphaLISA method can be used in clinical practice for the rapid, accurate, and sensitive detection of rotavirus infection.
Collapse
Affiliation(s)
- Ye Wang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
| | - Yuling Zheng
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
| | - Yan Li
- The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Shengwei Zhang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
- Department of Clinical Laboratory, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xin Wang
- The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Huijun Zong
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
- The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Wenhua Huang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
| | - Decong Kong
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
| | - Yongqiang Jiang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
| | - Peng Liu
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
- *Correspondence: Peng Liu
| | - Qingyu Lv
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
- Qingyu Lv
| | - Hua Jiang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
- Hua Jiang
| |
Collapse
|
8
|
Jin Y, He Y, Zhao D, Chen Y, Xue Q, Zou M, Yin H, Xing S. Development of an amplified luminescent proximity homogeneous assay for the detection of sulfonamides in animal-derived products. Food Sci Nutr 2021; 9:4938-4945. [PMID: 34532005 PMCID: PMC8441374 DOI: 10.1002/fsn3.2443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 12/02/2022] Open
Abstract
In this study, we carried out an amplified luminescent proximity homogeneous assay (AlphaLISA) to detect sulfonamides (SAs) antibiotic residues in plasma, milk, pork, chicken, and fish. The SAs AlphaLISA method can detect 13 SAs with half-inhibitory concentration (IC50) 2.11-29.77 ng/ml. The detection level of those SAs was 0.3-41.12 ng/ml in matrices, which satisfied the maximum residue limit (MRL) of the European Union, United States, and China. Our recoveries are in the range of 88% to 116.8% with a coefficient of variation less than 9.3% for different spiked food samples. We observed a good correlation between the AlphaLISA and liquid chromatography-tandem mass spectrometry (LC-MS/MS) with blood samples from injected rabbits. The established AlphaLISA method provided a no-washing, rapid, high-throughput screening tool for SAs in food quality control, which is suitable for small-volume samples.
Collapse
Affiliation(s)
- Yong Jin
- Chinese Academy of Inspection and QuarantineBeijingChina
| | - Yanping He
- Chinese Academy of Inspection and QuarantineBeijingChina
- Anhui Normal UniversityWuhuChina
| | - Dali Zhao
- Jilin International Travel Health Care Center (Changchun Customs Port Clinic)ChangchunChina
| | - Yan Chen
- Chinese Academy of Inspection and QuarantineBeijingChina
| | - Qiang Xue
- Chinese Academy of Inspection and QuarantineBeijingChina
| | - Mingqiang Zou
- Chinese Academy of Inspection and QuarantineBeijingChina
| | - Hong Yin
- Chinese Academy of Inspection and QuarantineBeijingChina
| | - Shige Xing
- Chinese Academy of Inspection and QuarantineBeijingChina
| |
Collapse
|
9
|
Ramadan Q, Fardous RS, Hazaymeh R, Alshmmari S, Zourob M. Pharmacokinetics-On-a-Chip: In Vitro Microphysiological Models for Emulating of Drugs ADME. Adv Biol (Weinh) 2021; 5:e2100775. [PMID: 34323392 DOI: 10.1002/adbi.202100775] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 06/08/2021] [Indexed: 12/15/2022]
Abstract
Despite many ongoing efforts across the full spectrum of pharmaceutical and biotech industries, drug development is still a costly undertaking that involves a high risk of failure during clinical trials. Animal models played vital roles in understanding the mechanism of human diseases. However, the use of these models has been a subject of heated debate, particularly due to ethical matters and the inevitable pathophysiological differences between animals and humans. Current in vitro models lack the sufficient functionality and predictivity of human pharmacokinetics and toxicity, therefore, are not capable to fully replace animal models. The recent development of micro-physiological systems has shown great potential as indispensable tools for recapitulating key physiological parameters of humans and providing in vitro methods for predicting the pharmacokinetics and pharmacodynamics in humans. Integration of Absorption, Distribution, Metabolism, and Excretion (ADME) processes within one close in vitro system is a paramount development that would meet important unmet pharmaceutical industry needs. In this review paper, synthesis of the ADME-centered organ-on-a-chip technology is systemically presented from what is achieved to what needs to be done, emphasizing the requirements of in vitro models that meet industrial needs in terms of the structure and functions.
Collapse
Affiliation(s)
- Qasem Ramadan
- Alfaisal University, Riyadh, 11533, Kingdom of Saudi Arabia
| | - Roa Saleem Fardous
- Alfaisal University, Riyadh, 11533, Kingdom of Saudi Arabia.,Strathclyde Institute of Pharmacy and Biomedical Sciences, Strathclyde University, Glasgow, G4 0RE, United Kingdom
| | - Rana Hazaymeh
- Almaarefa University, Riyadh, 13713, Kingdom of Saudi Arabia
| | - Sultan Alshmmari
- Saudi Food and Drug Authority, Riyadh, 13513-7148, Kingdom of Saudi Arabia
| | | |
Collapse
|
10
|
Zheng Y, Shao Y, Fu J. A microfluidics-based stem cell model of early post-implantation human development. Nat Protoc 2020; 16:309-326. [PMID: 33311712 DOI: 10.1038/s41596-020-00417-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
Early post-implantation human embryonic development has been challenging to study due to both technical limitations and ethical restrictions. Proper modeling of the process is important for infertility and toxicology research. Here we provide details of the design and implementation of a microfluidic device that can be used to model human embryo development. The microfluidic human embryo model is established from human pluripotent stem cells (hPSCs), and the resulting structures exhibit molecular and cellular features resembling the progressive development of the early post-implantation human embryo. The compartmentalized configuration of the microfluidic device allows the formation of spherical hPSC clusters in prescribed locations in the device, enabling the two opposite regions of each hPSC cluster to be exposed to two different exogenous chemical environments. Under such asymmetrical chemical conditions, several early post-implantation human embryo developmental landmarks, including lumenogenesis of the epiblast and the resultant pro-amniotic cavity, formation of a bipolar embryonic sac, and specification of primordial germ cells and gastrulating cells (or mesendoderm cells), can be robustly recapitulated using the microfluidic device. The microfluidic human embryo model is compatible with high-throughput studies, live imaging, immunofluorescence staining, fluorescent in situ hybridization, and single-cell sequencing. This protocol takes ~5 d to complete, including microfluidic device fabrication (2 d), cell seeding (1 d), and progressive development of the microfluidic model until gastrulation-like events occur (1-2 d).
Collapse
Affiliation(s)
- Yi Zheng
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Yue Shao
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA.,Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, School of Aerospace Engineering, Tsinghua University, Beijing, China
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA. .,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA. .,Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
11
|
Abdullah MAA, Amini N, Yang L, Paluh JL, Wang J. Multiplexed analysis of neural cytokine signaling by a novel neural cell-cell interaction microchip. LAB ON A CHIP 2020; 20:3980-3995. [PMID: 32945325 PMCID: PMC7606659 DOI: 10.1039/d0lc00401d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Multipotent neural stem cells (NSCs) are widely applied in pre-clinical and clinical trials as a cell source to promote tissue regeneration in neurodegenerative diseases. Frequently delivered as dissociated cells, aggregates or self-organized rosettes, it is unknown whether disruption of the NSC rosette morphology or method of formation affect signaling profiles of these cells that may impact uniformity of outcomes in cell therapies. Here we generate a neural cell-cell interaction microchip (NCCIM) as an in vitro platform to simultaneously track an informed panel of cytokines and co-evaluate cell morphology and biomarker expression coupled to a sandwich ELISA platform. We apply multiplex in situ tagging technology (MIST) to evaluate ten cytokines (PDGF-AA, GDNF, BDNF, IGF-1, FGF-2, IL-6, BMP-4, CNTF, β-NGF, NT-3) on microchips for EB-derived rosettes, single cell dissociated rosettes and reformed rosette neurospheres. Of the cytokines evaluated, EB-derived rosettes secrete PDGF-AA, GDNF and FGF-2 prominently, whereas this profile is temporarily lost upon dissociation to single cells and in reformed neurospheres two additional cytokines, BDNF and β-NGF, are also secreted. This study on NSC rosettes demonstrates the development, versatility and utility of the NCCIM as a sensitive multiplex detector of cytokine signaling in a high throughput and controlled microenvironment. The NCCIM is expected to provide important new information to refine cell source choices in therapies as well as to support development of informative 2D or 3D in vitro models including areas of neurodegeneration or neuroplasticity.
Collapse
Affiliation(s)
- Mohammed A. A. Abdullah
- Multiplex Biotechnology Laboratory, Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794
- Department of Chemistry, State University of New York at Albany, Albany, NY 12222
| | - Nooshin Amini
- Nanobioscience, State University of New York Polytechnic Institute, Albany, NY 12203
| | - Liwei Yang
- Multiplex Biotechnology Laboratory, Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794
| | - Janet L. Paluh
- Nanobioscience, State University of New York Polytechnic Institute, Albany, NY 12203
- Corresponding authors. ;
| | - Jun Wang
- Multiplex Biotechnology Laboratory, Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794
- Corresponding authors. ;
| |
Collapse
|
12
|
Sinibaldi A, Doricchi A, Pileri T, Allegretti M, Danz N, Munzert P, Giordani E, Giacomini P, Michelotti F. Bioassay engineering: a combined label-free and fluorescence approach to optimize HER2 detection in complex biological media. Anal Bioanal Chem 2020; 412:3509-3517. [PMID: 32300843 DOI: 10.1007/s00216-020-02643-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/03/2020] [Indexed: 01/16/2023]
Abstract
We report on the combined label-free/fluorescence use of one-dimensional photonic crystals to optimize cancer biomarker detection in complex biological media. The optimization of the assay working parameters permits us to maximize the final response of the biosensor. The detection approach utilizes a sandwich assay, in which one-dimensional photonic crystals sustaining Bloch surface waves are modified with monoclonal antibodies in order to guarantee high specificity during biological recognition. The multiple outcomes generated by such optimization experiments permitted us to determine the effective capture efficiency and the repeatability of the immobilization process, which was estimated to be close to 5%. By exploiting the resolution of the fluorescence operation mode, we studied non-specific interactions in different blocking agents, different analyte diluting buffers, and diverse concentrations of the detection antibody. As a clinically relevant biomarker, we selected the trans-membrane receptor tyrosine kinase HER2. HER2 regulates a variety of cell proliferation, growth, and differentiation pathways and its over-expression occurs in approximately 20-30% of breast cancer worldwide. As a final application, we transferred all the optimized working parameters to HER2 cancer biomarker assays in a complex biological environment. The label-free and fluorescence results obtained by analyzing MCF-7 (HER2 low positive) and 32D (HER2 negative) cell lysates demonstrate that we can successfully discriminate the two lysates.
Collapse
Affiliation(s)
- Alberto Sinibaldi
- Department of Basic and Applied Science for Engineering, Sapienza University of Rome, Via A. Scarpa 16, 00161, Rome, Italy.
| | - Andrea Doricchi
- Department of Basic and Applied Science for Engineering, Sapienza University of Rome, Via A. Scarpa 16, 00161, Rome, Italy
| | - Tommaso Pileri
- Department of Basic and Applied Science for Engineering, Sapienza University of Rome, Via A. Scarpa 16, 00161, Rome, Italy
| | - Matteo Allegretti
- IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Norbert Danz
- Fraunhofer Institute for Applied Optics and Engineering IOF, A. Einstein-Str. 7, 07745, Jena, Germany
| | - Peter Munzert
- Fraunhofer Institute for Applied Optics and Engineering IOF, A. Einstein-Str. 7, 07745, Jena, Germany
| | - Elena Giordani
- IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Patrizio Giacomini
- IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Francesco Michelotti
- Department of Basic and Applied Science for Engineering, Sapienza University of Rome, Via A. Scarpa 16, 00161, Rome, Italy
| |
Collapse
|
13
|
Guo Q, Wang Y, Chen C, Wei D, Fu J, Xu H, Gu H. Multiplexed Luminescence Oxygen Channeling Immunoassay Based on Dual-Functional Barcodes with a Host-Guest Structure: A Facile and Robust Suspension Array Platform. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1907521. [PMID: 32174029 DOI: 10.1002/smll.201907521] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 02/08/2020] [Accepted: 02/19/2020] [Indexed: 05/24/2023]
Abstract
The development of a powerful immunoassay platform with capacities of both simplicity and high multiplexing is promising for disease diagnosis. To meet this urgent need, for the first time, a multiplexed luminescent oxygen channeling immunoassay (multi-LOCI) platform by implementation of LOCI with suspension array technology is reported. As the microcarrier of the platform, a unique dual-functional barcode with a host-guest structure composed of a quantum dot host bead (QDH) and LOCI acceptor beads (ABs) is designed, in which QDH provides function of high coding capacity while ABs facilitate the LOCI function. The analytes bridge QDH@ABs and LOCI donor beads (DBs) into a close proximity, forming a QDH@ABs-DBs "host-guest-satellite" superstructure that generates both barcode signal from QDH and LOCI signal induced by singlet oxygen channeling between ABs and DBs. Through imaging-based decoding, different barcodes are automatically distinguished and colocalized with LOCI signals. Importantly, the assay achieves simultaneous detection of multiple analytes within one reaction, simply by following a "mix-and-measure" protocol without the need for tedious washing steps. Furthermore, the multi-LOCI platform is validated for real sample measurements. With the advantages of robustness, simplicity, and high multiplexing, the platform holds great potential for the development of point-of-care diagnostics.
Collapse
Affiliation(s)
- Qingsheng Guo
- Shanghai Jiao Tong University Affiliated Sixth Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Yao Wang
- Shanghai Jiao Tong University Affiliated Sixth Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Cang Chen
- Shanghai Jiao Tong University Affiliated Sixth Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Dan Wei
- Shanghai Jiao Tong University Affiliated Sixth Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Jianping Fu
- Department of Mechanical Engineering, Department of Biomedical Engineering, Department of Cell and Developmental Biology, University of Michiga Ann Arbor, Ann Arbor, MI, 48109, USA
| | - Hong Xu
- Shanghai Jiao Tong University Affiliated Sixth Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Hongchen Gu
- Shanghai Jiao Tong University Affiliated Sixth Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| |
Collapse
|
14
|
Liu J, Abdullah MAA, Yang L, Wang J. Fast Affinity Induced Reaction Sensor Based on a Fluorogenic Click Reaction for Quick Detection of Protein Biomarkers. Anal Chem 2019; 92:647-653. [PMID: 31790589 DOI: 10.1021/acs.analchem.9b04502] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Despite numerous biosensors currently available, the routine biomarker detection still largely relies on traditional ELISA and Western blot. Those standard techniques are labor intensive and time-consuming. Herein we introduce a fast affinity induced reaction sensor (FAIRS) that overcomes a few limitations of traditional and emerging biosensors. FAIRS is a general, one-step method and is naturally specific in detection. FAIRS probes are composed of a sandwich ELISA antibody pair that is conjugated with two fluorogenic click chemicals. This technology leverages significant differences of antibody affinity and chemical reaction rate, which are characterized to guide probe design. The stability, sensitivity, detection range, and response time are fully characterized. Application to IL-6 detection using blood serum and cell culture medium demonstrates that FAIRS can quantify IL-6 with high sensitivity in one step. With the unique features, FAIRS probes may find broad applications in medical sciences and clinical diagnostics, where quick detection of biomarkers is demanded.
Collapse
Affiliation(s)
- Jingxin Liu
- Multiplex Biotechnology Laboratory, Department of Biomedical Engineering , State University of New York at Stony Brook , Stony Brook , New York 11788 , United States
| | - Mohammed A A Abdullah
- Multiplex Biotechnology Laboratory, Department of Biomedical Engineering , State University of New York at Stony Brook , Stony Brook , New York 11788 , United States.,Department of Chemistry , State University of New York, University at Albany , Albany , New York 12222 , United States
| | - Liwei Yang
- Multiplex Biotechnology Laboratory, Department of Biomedical Engineering , State University of New York at Stony Brook , Stony Brook , New York 11788 , United States
| | - Jun Wang
- Multiplex Biotechnology Laboratory, Department of Biomedical Engineering , State University of New York at Stony Brook , Stony Brook , New York 11788 , United States
| |
Collapse
|
15
|
Nguyen HV, Nguyen VD, Nguyen HQ, Chau THT, Lee EY, Seo TS. Nucleic acid diagnostics on the total integrated lab-on-a-disc for point-of-care testing. Biosens Bioelectron 2019; 141:111466. [DOI: 10.1016/j.bios.2019.111466] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/14/2019] [Accepted: 06/21/2019] [Indexed: 12/15/2022]
|
16
|
Dang BV, Hassanzadeh-Barforoushi A, Syed MS, Yang D, Kim SJ, Taylor RA, Liu GJ, Liu G, Barber T. Microfluidic Actuation via 3D-Printed Molds toward Multiplex Biosensing of Cell Apoptosis. ACS Sens 2019; 4:2181-2189. [PMID: 31321976 DOI: 10.1021/acssensors.9b01057] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Multiplexed analysis of biochemical analytes such as proteins, enzymes, and immune products using a microfluidic device has the potential to cut assay time, reduce sample volume, realize high-throughput, and decrease experimental error without compromising sensitivity. Despite these huge benefits, the need for expensive specialized equipment and the complex photolithography fabrication process for the multiplexed devices have, to date, prevented widespread adoption of microfluidic systems. Here, we present a simple method to fabricate a new microfluidic-based multiplexed biosensing device by taking advantage of 3D-printing. The device is an integration of normally closed (NC) microfluidic valving units which offer superior operational flexibility by using PDMS membrane (E ∼ 1-2 MPa) and require minimized energy input (1-5 kPa). To systematically engineer the device, we first report on the geometrical and operational analysis of a single 3D-printed valving unit. Based on the characterization, we introduce a full prototype multiplexed chip comprising several microfluidic valves. The prototype offers-for the first time in a 3D-printed microfluidic device-the capability of on-demand performce of both a sequential and a parallel biochemical assay. As a proof of concept, our device has been used to simultaneously measure the apoptotic activity of 5 different members of the caspase protease enzyme family. In summary, the 3D-printed valving system showcased in this study overcomes traditional bottlenecks of microfabrication, enabling a new class of sophisticated liquid manipulation required in performing multiplexed sensing for biochemical assays.
Collapse
Affiliation(s)
- Bac Van Dang
- School of Mechanical and Manufacturing Engineering, Faculty of Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Amin Hassanzadeh-Barforoushi
- School of Mechanical and Manufacturing Engineering, Faculty of Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia
- Cancer Division, Garvan Institute of Medical Research/the Kinghorn Cancer Centre, Sydney, New South Wales 2010, Australia
| | - Maira Shakeel Syed
- School of Mechanical and Manufacturing Engineering, Faculty of Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Danting Yang
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale BioPhotonics, Australian Centre for NanoMedicine, Faculty of Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Medical School of Ningbo University, Ningbo, Zhejiang 315211, China
| | - Sung-Jin Kim
- Department of Mechanical Engineering, Konkuk University, Seoul, 05029, Republic of Korea
| | - Robert A. Taylor
- School of Mechanical and Manufacturing Engineering, Faculty of Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Guo-Jun Liu
- Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, New South Wales 2234, Australia
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Medicine and Health, Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, New South Wales 2050, Australia
| | - Guozhen Liu
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale BioPhotonics, Australian Centre for NanoMedicine, Faculty of Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Tracie Barber
- School of Mechanical and Manufacturing Engineering, Faculty of Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
17
|
Zhao J, Lv Q, Liu P, Guo L, Zhang L, Zheng Y, Ming L, Kong D, Jiang H, Jiang Y. AlphaLISA for detection of staphylococcal enterotoxin B free from interference by protein A. Toxicon 2019; 165:62-68. [DOI: 10.1016/j.toxicon.2019.04.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 04/03/2019] [Accepted: 04/24/2019] [Indexed: 01/15/2023]
|
18
|
Kurmashev A, Kwon S, Park JK, Kang JH. Vertically sheathing laminar flow-based immunoassay using simultaneous diffusion-driven immune reactions. RSC Adv 2019; 9:23791-23796. [PMID: 35530621 PMCID: PMC9069447 DOI: 10.1039/c9ra03855h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 07/24/2019] [Indexed: 11/21/2022] Open
Abstract
Simultaneous infusion of primary and secondary antibodies of different diffusivity into vertical laminar flows enables the improved immune reactions.
Collapse
Affiliation(s)
- Amanzhol Kurmashev
- Department of Biomedical Engineering
- School of Life Sciences
- Ulsan National Institute of Science and Technology (UNIST)
- Ulsan
- Republic of Korea
| | - Seyong Kwon
- Department of Biomedical Engineering
- School of Life Sciences
- Ulsan National Institute of Science and Technology (UNIST)
- Ulsan
- Republic of Korea
| | - Je-Kyun Park
- Department of Bio and Brain Engineering
- Korea Advanced Institute of Science and Technology (KAIST)
- Daejeon
- Republic of Korea
| | - Joo H. Kang
- Department of Biomedical Engineering
- School of Life Sciences
- Ulsan National Institute of Science and Technology (UNIST)
- Ulsan
- Republic of Korea
| |
Collapse
|
19
|
Yang Y, Zeng Y. Microfluidic communicating vessel chip for expedited and automated immunomagnetic assays. LAB ON A CHIP 2018; 18:3830-3839. [PMID: 30394473 PMCID: PMC6279511 DOI: 10.1039/c8lc00927a] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Rapid, sensitive analysis of protein biomarkers is of tremendous biological and clinical significance. Immunoassays are workhorse tools for protein analysis and have been under continuous investigation to develop new methods and to improve the analytical performance. Herein we report a pneumatically gated microfluidic communicating vessel (μCOVE) chip for rapid and sensitive immunomagnetic ELISA. A distinct feature of our device is that it employs the communicating vessel principle as a simple means to generate a fast transient hydrodynamic flow to enable effective flow washing without the need for excessive incubation, which greatly simplifies and expedites the assay workflow, compared to conventional microfluidic flow-based immunoassays. Stationary multi-phase microfluidic techniques have been developed for fast bead washing. However, they have some limitations, such as the need for careful control of interfacial properties, large bead quantity required for reliable interphase bead transport, and relatively high bead loss during surface tension-gated traverse. Our single-phase μCOVE chip can overcome such limitations and facilitate the manipulation of magnetic beads to streamline the assay workflow. We showed that the μCOVE device affords highly sensitive quantification of the CEA and EGFR proteins with a LOD down to the sub-picogram per mL level. Direct detection of the EGFR in the crude A431 cell lysate was also demonstrated to further validate the ability of our device for rapid and quantitative analysis of complex biological samples. Overall, our work presents a unique platform that combines the merits of the stationary multi-phase systems and the flow-based microfluidics. This novel immunoassay microsystem has promising potential for a broad range of biological and clinical applications, owing to its simplicity and high performance.
Collapse
Affiliation(s)
- Yang Yang
- Department of Chemistry, University of Kansas, Lawrence, KS 66045, USA.
| | | |
Collapse
|
20
|
Pan Q, Yamauchi KA, Herr AE. Controlling Dispersion during Single-Cell Polyacrylamide-Gel Electrophoresis in Open Microfluidic Devices. Anal Chem 2018; 90:13419-13426. [PMID: 30346747 PMCID: PMC6777840 DOI: 10.1021/acs.analchem.8b03233] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
New tools for measuring protein expression in individual cells complement single-cell genomics and transcriptomics. To characterize a population of individual mammalian cells, hundreds to thousands of microwells are arrayed on a polyacrylamide-gel-coated glass microscope slide. In this "open" fluidic device format, we explore the feasibility of mitigating diffusional losses during lysis and polyacrylamide-gel electrophoresis (PAGE) through spatial control of the pore-size of the gel layer. To reduce in-plane diffusion-driven dilution of each single-cell lysate during in-microwell chemical lysis, we photopattern and characterize microwells with small-pore-size sidewalls ringing the microwell except at the injection region. To reduce out-of-plane-diffusion-driven-dilution-caused signal loss during both lysis and single-cell PAGE, we scrutinize a selectively permeable agarose lid layer. To reduce injection dispersion, we photopattern and study a stacking-gel feature at the head of each <1 mm separation axis. Lastly, we explore a semienclosed device design that reduces the cross-sectional area of the chip, thus reducing Joule-heating-induced dispersion during single-cell PAGE. As a result, we observed a 3-fold increase in separation resolution during a 30 s separation and a >2-fold enhancement of the signal-to-noise ratio. We present well-integrated strategies for enhancing overall single-cell-PAGE performance.
Collapse
Affiliation(s)
- Qiong Pan
- Department of Bioengineering, University of California, Berkeley, California 94720, United States
| | - Kevin A. Yamauchi
- Department of Bioengineering, University of California, Berkeley, California 94720, United States
| | - Amy E. Herr
- Department of Bioengineering, University of California, Berkeley, California 94720, United States
| |
Collapse
|
21
|
Dixit C, Kadimisetty K, Rusling J. 3D-printed miniaturized fluidic tools in chemistry and biology. Trends Analyt Chem 2018; 106:37-52. [PMID: 32296252 PMCID: PMC7158885 DOI: 10.1016/j.trac.2018.06.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
3D printing (3DP), an additive manufacturing (AM) approach allowing for rapid prototyping and decentralized fabrication on-demand, has become a common method for creating parts or whole devices. The wide scope of the AM extends from organized sectors of construction, ornament, medical, and R&D industries to individual explorers attributed to the low cost, high quality printers along with revolutionary tools and polymers. While progress is being made but big manufacturing challenges are still there. Considering the quickly shifting narrative towards miniaturized analytical systems (MAS) we focus on the development/rapid prototyping and manufacturing of MAS with 3DP, and application dependent challenges in engineering designs and choice of the polymeric materials and provide an exhaustive background to the applications of 3DP in biology and chemistry. This will allow readers to perceive the most important features of AM in creating (i) various individual and modular components, and (ii) complete integrated tools.
Collapse
Affiliation(s)
- C.K. Dixit
- Department of Chemistry, University of Connecticut, Storrs, CT 06269-3060, United States
| | - K. Kadimisetty
- Department of Chemistry, University of Connecticut, Storrs, CT 06269-3060, United States
| | - J. Rusling
- Department of Chemistry, University of Connecticut, Storrs, CT 06269-3060, United States
- Institute of Materials Science, University of Connecticut, Storrs, CT 06269-3136, United States
- Department of Surgery and Neag Cancer Centre, UConn Health, Farmington, CT 06030, United States
- School of Chemistry, National University of Ireland at Galway, Galway, Ireland
| |
Collapse
|
22
|
Geng S, Huang Y. From Mouth Pipetting to Microfluidics: The Evolution of Technologies for Picking Healthy Single Cells. ACTA ACUST UNITED AC 2018. [DOI: 10.1002/adbi.201800099] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Shuang Geng
- Beijing Advanced Innovation Center for Genomics (ICG); Biodynamics Optical Imaging Center (BIOPIC); School of Life Sciences; Peking-Tsinghua Center for Life Sciences; and College of Engineering; Peking University; Beijing 100871 China
| | - Yanyi Huang
- Beijing Advanced Innovation Center for Genomics (ICG); Biodynamics Optical Imaging Center (BIOPIC); School of Life Sciences; Peking-Tsinghua Center for Life Sciences; and College of Engineering; Peking University; Beijing 100871 China
| |
Collapse
|
23
|
Ubillos I, Aguilar R, Sanz H, Jiménez A, Vidal M, Valmaseda A, Dong Y, Gaur D, Chitnis CE, Dutta S, Angov E, Aponte JJ, Campo JJ, Valim C, Harezlak J, Dobaño C. Analysis of factors affecting the variability of a quantitative suspension bead array assay measuring IgG to multiple Plasmodium antigens. PLoS One 2018; 13:e0199278. [PMID: 29966018 PMCID: PMC6028107 DOI: 10.1371/journal.pone.0199278] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 05/28/2018] [Indexed: 01/25/2023] Open
Abstract
Reducing variability of quantitative suspension array assays is key for multi-center and large sero-epidemiological studies. To maximize precision and robustness of an in-house IgG multiplex assay, we analyzed the effect of several conditions on variability to find the best combination. The following assay conditions were studied through a fractional factorial design: antigen-bead coupling (stock vs. several), sample predilution (stock vs. daily), temperature of incubation of sample with antigen-bead (22°C vs. 37°C), plate washing (manual vs. automatic) and operator expertise (expert vs. apprentice). IgG levels against seven P. falciparum antigens with heterogeneous immunogenicities were measured in test samples, in a positive control and in blanks. We assessed the variability and MFI quantification range associated to each combination of conditions, and their interactions, and evaluated the minimum number of samples and blank replicates to achieve good replicability. Results showed that antigen immunogenicity and sample seroreactivity defined the optimal dilution to assess the effect of assay conditions on variability. We found that a unique antigen-bead coupling, samples prediluted daily, incubation at 22°C, and automatic washing, had lower variability. However, variability increased when performing several couplings and incubating at 22°C vs. 37°C. In addition, no effect of temperature was seen with a unique coupling. The expertise of the operator had no effect on assay variability but reduced the MFI quantification range. Finally, differences between sample replicates were minimal, and two blanks were sufficient to capture assay variability, as suggested by the constant Intraclass Correlation Coefficient of three and two blanks. To conclude, a single coupling was the variable that most consistently reduced assay variability, being clearly advisable. In addition, we suggest having more sample dilutions instead of replicates to increase the likelihood of sample MFIs falling in the linear part of the antigen-specific curve, thus increasing precision.
Collapse
Affiliation(s)
- Itziar Ubillos
- ISGlobal, Hospital Clínic—Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Ruth Aguilar
- ISGlobal, Hospital Clínic—Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Hector Sanz
- ISGlobal, Hospital Clínic—Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Alfons Jiménez
- ISGlobal, Hospital Clínic—Universitat de Barcelona, Barcelona, Catalonia, Spain
- CIBER Epidemiology and Public Health (CIBERESP), Barcelona, Spain
| | - Marta Vidal
- ISGlobal, Hospital Clínic—Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Aida Valmaseda
- ISGlobal, Hospital Clínic—Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Yan Dong
- Department of Biostatistics, RM Fairbanks School of Public Health, Indianapolis, IN, United States of America
| | - Deepak Gaur
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Chetan E. Chitnis
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Sheetij Dutta
- U.S. Military Malaria Vaccine Program, Walter Reed Army Institute of Research (WRAIR), Silver Spring, Maryland, United States of America
| | - Evelina Angov
- U.S. Military Malaria Vaccine Program, Walter Reed Army Institute of Research (WRAIR), Silver Spring, Maryland, United States of America
| | - John J. Aponte
- ISGlobal, Hospital Clínic—Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Joseph J. Campo
- ISGlobal, Hospital Clínic—Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Clarissa Valim
- Department of Osteopathic Medical Specialties, Michigan State University, East Lansing, MI, United States of America
- Department of Immunology and Infectious Diseases, Harvard T.H. Chen School of Public Health, Boston, MA, United States of America
| | - Jaroslaw Harezlak
- Department of Biostatistics, RM Fairbanks School of Public Health, Indianapolis, IN, United States of America
| | - Carlota Dobaño
- ISGlobal, Hospital Clínic—Universitat de Barcelona, Barcelona, Catalonia, Spain
- * E-mail:
| |
Collapse
|
24
|
Kadimisetty K, Malla S, Bhalerao KS, Mosa IM, Bhakta S, Lee NH, Rusling JF. Automated 3D-Printed Microfluidic Array for Rapid Nanomaterial-Enhanced Detection of Multiple Proteins. Anal Chem 2018; 90:7569-7577. [PMID: 29779368 PMCID: PMC6104517 DOI: 10.1021/acs.analchem.8b01198] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We report here the fabrication and validation of a novel 3D-printed, automated immunoarray to detect multiple proteins with ultralow detection limits. This low cost, miniature immunoarray employs electrochemiluminescent (ECL) detection measured with a CCD camera and employs touch-screen control of a micropump to facilitate automated use. The miniaturized array features prefilled reservoirs to deliver sample and reagents to a paper-thin pyrolytic graphite microwell detection chip to complete sandwich immunoassays. The detection chip achieves high sensitivity by using single-wall carbon nanotube-antibody conjugates in the microwells and employing massively labeled antibody-decorated RuBPY-silica nanoparticles to generate ECL. The total cost of an array is $0.65, and an eight-protein assay can be done in duplicate for $0.14 per protein with limits of detection (LOD) as low as 78-110 fg mL-1 in diluted serum. The electronic control system costs $210 in components. Utility of the automated immunoarray was demonstrated by detecting an eight-protein prostate cancer biomarker panel in human serum samples in 25 min. The system is well suited to future clinical and point-of-care diagnostic testing and could be used in resource-limited environments.
Collapse
Affiliation(s)
- Karteek Kadimisetty
- Department of Chemistry and Institute of Material Science, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Spundana Malla
- Department of Chemistry and Institute of Material Science, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Ketki S. Bhalerao
- Department of Chemistry and Institute of Material Science, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Islam M. Mosa
- Department of Chemistry and Institute of Material Science, University of Connecticut, Storrs, Connecticut 06269, United States
- Department of Chemistry, Tanta University, Tanta 31527, Egypt
| | - Snehasis Bhakta
- Department of Chemistry and Institute of Material Science, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Norman H. Lee
- Department of Pharmacology & Physiology, George Washington University, Washington, D.C. 20037, United States
| | - James F. Rusling
- Department of Chemistry and Institute of Material Science, University of Connecticut, Storrs, Connecticut 06269, United States
- Institute of Material Science, University of Connecticut, Storrs, Connecticut 06269, United States
- Department of Surgery and Neag Cancer Center, UConn Health, Farmington, Connecticut 06032, United States
- School of Chemistry, National University of Ireland, Galway H91 TK33, Ireland
| |
Collapse
|
25
|
Veenuttranon K, Nguyen LT. Programmable electrochemical flow system for high throughput determination of total antioxidant capacity. Talanta 2018; 186:286-292. [PMID: 29784362 DOI: 10.1016/j.talanta.2018.04.073] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 04/22/2018] [Accepted: 04/23/2018] [Indexed: 01/07/2023]
Abstract
The aim of this work was to develop a programmable flow system for rapid assessment of total antioxidant capacity (TAC). Novel features of the prototype include a single pressure-driven source, versatile manipulation of fluid flows, ability to adapt to different TAC assays, and compatibility with microfluidic design. Antioxidant activity was determined by electrochemical measurement of residual 2,2-diphenyl-2-picrylhydrayl hydrate (DPPH•) free radicals in the solution. The overall performance of the device was validated by the spectrophotometric method. The results indicated that dosing of reagents and samples could be controlled by pressure (R2 = 0.992) and time (R2 = 0.999) with high accuracy, and the mixing uniformity of the device was equivalent to that of the batch mixing (R2 = 0.994). TAC assays of a standard antioxidant, ascorbic acid, as well as selected samples such as orange and pomegranate juices, white wine, and green tea by the device were comparable to traditional measurement techniques. Due to the short incubation time, the approach may be more suitable for fast, rather than slow reacting antioxidant compounds. The developed system could be used for rapid TAC screening of food and biological samples.
Collapse
Affiliation(s)
- Kornautchaya Veenuttranon
- Department of Food, Agriculture and Bioresources, School of Environment, Resources and Development, Asian Institute of Technology, 58 Moo 9, Km. 42, Paholyothin Highway, Klong Luang, Pathumthani 12120, Thailand
| | - Loc Thai Nguyen
- Department of Food, Agriculture and Bioresources, School of Environment, Resources and Development, Asian Institute of Technology, 58 Moo 9, Km. 42, Paholyothin Highway, Klong Luang, Pathumthani 12120, Thailand.
| |
Collapse
|
26
|
Yan L, Dong X, Gao J, Liu F, Zhou L, Sun Y, Zhao X. A novel rapid quantitative method reveals stathmin-1 as a promising marker for esophageal squamous cell carcinoma. Cancer Med 2018; 7:1802-1813. [PMID: 29577639 PMCID: PMC5943482 DOI: 10.1002/cam4.1449] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 02/08/2018] [Accepted: 02/25/2018] [Indexed: 12/18/2022] Open
Abstract
Stathmin‐1 is a microtubule depolymerization protein that regulates cell division, growth, migration, and invasion. Overexpression of stathmin‐1 has been observed to be associated with metastasis, poor prognosis, and chemoresistance in various human cancers. Our previous studies found that serum stathmin‐1 was significantly elevated in patients with esophageal squamous cell carcinoma (ESCC) by ELISAs. Here, we constructed high‐affinity monoclonal antibodies and then developed a competitive AlphaLISA for rapid, accurate quantitation of stathmin‐1 in serum. Compared to ELISA, our homogeneous AlphaLISA showed better sensitivity and accuracy, a lower limit of detection, and a wider linear range. The measurements of nearly 1000 clinical samples showed that serum stathmin‐1 level increased dramatically in patients with squamous cell carcinoma (SCC), especially in ESCC, with a sensitivity and a specificity of 81% and 94%, respectively. Even for early stage ESCC, stathmin‐1 achieved an area under the receiver operating characteristic curve (AUC) of 0.88. Meanwhile, raised concentrations of stathmin‐1 were associated with lymph node metastasis and advanced cancer stage. Notably, various types of SCC showed significantly higher AUCs in serum stathmin‐1 detection compared to adenocarcinoma. Furthermore, we confirmed that stathmin‐1 was enriched in the oncogenic exosomes, which can explain the reason why it enters into the blood to serve as a tumor surrogate. In conclusion, this large‐scale and systematic study of serum stathmin‐1 measured by our newly established AlphaLISA showed that stathmin‐1 is a very promising diagnostic and predictive marker for SCC in the clinic, especially for ESCC.
Collapse
Affiliation(s)
- Lu Yan
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiu Dong
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiajia Gao
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fang Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lanping Zhou
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yulin Sun
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaohang Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
27
|
Shrirao AB, Fritz Z, Novik EM, Yarmush GM, Schloss RS, Zahn JD, Yarmush ML. Microfluidic flow cytometry: The role of microfabrication methodologies, performance and functional specification. TECHNOLOGY 2018; 6:1-23. [PMID: 29682599 PMCID: PMC5907470 DOI: 10.1142/s2339547818300019] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Flow cytometry is an invaluable tool utilized in modern biomedical research and clinical applications requiring high throughput, high resolution particle analysis for cytometric characterization and/or sorting of cells and particles as well as for analyzing results from immunocytometric assays. In recent years, research has focused on developing microfluidic flow cytometers with the motivation of creating smaller, less expensive, simpler, and more autonomous alternatives to conventional flow cytometers. These devices could ideally be highly portable, easy to operate without extensive user training, and utilized for research purposes and/or point-of-care diagnostics especially in limited resource facilities or locations requiring on-site analyses. However, designing a device that fulfills the criteria of high throughput analysis, automation and portability, while not sacrificing performance is not a trivial matter. This review intends to present the current state of the field and provide considerations for further improvement by focusing on the key design components of microfluidic flow cytometers. The recent innovations in particle focusing and detection strategies are detailed and compared. This review outlines performance matrix parameters of flow cytometers that are interdependent with each other, suggesting trade offs in selection based on the requirements of the applications. The ongoing contribution of microfluidics demonstrates that it is a viable technology to advance the current state of flow cytometry and develop automated, easy to operate and cost-effective flow cytometers.
Collapse
Affiliation(s)
- Anil B Shrirao
- Department of Biomedical Engineering, Rutgers University, 599, Taylor Road, Piscataway, NJ 08854
| | - Zachary Fritz
- Department of Biomedical Engineering, Rutgers University, 599, Taylor Road, Piscataway, NJ 08854
| | - Eric M Novik
- Hurel Corporation, 671, Suite B, U.S. Highway 1, North Brunswick, NJ 08902
| | - Gabriel M Yarmush
- Department of Biomedical Engineering, Rutgers University, 599, Taylor Road, Piscataway, NJ 08854
| | - Rene S Schloss
- Department of Biomedical Engineering, Rutgers University, 599, Taylor Road, Piscataway, NJ 08854
| | - Jeffrey D Zahn
- Department of Biomedical Engineering, Rutgers University, 599, Taylor Road, Piscataway, NJ 08854
| | - Martin L Yarmush
- Department of Biomedical Engineering, Rutgers University, 599, Taylor Road, Piscataway, NJ 08854
| |
Collapse
|
28
|
Parween S, Singh G, Nahar P. Ultrafast image-based ELISA for sensitive detection of cytokines in allergen-induced asthmatic samples. Microchem J 2017. [DOI: 10.1016/j.microc.2017.07.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
29
|
Senga Y, Imamura H, Miyafusa T, Watanabe H, Honda S. AlphaScreen-based homogeneous assay using a pair of 25-residue artificial proteins for high-throughput analysis of non-native IgG. Sci Rep 2017; 7:12466. [PMID: 28963557 PMCID: PMC5622108 DOI: 10.1038/s41598-017-12693-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 09/18/2017] [Indexed: 12/24/2022] Open
Abstract
Therapeutic IgG becomes unstable under various stresses in the manufacturing process. The resulting non-native IgG molecules tend to associate with each other and form aggregates. Because such aggregates not only decrease the pharmacological effect but also become a potential risk factor for immunogenicity, rapid analysis of aggregation is required for quality control of therapeutic IgG. In this study, we developed a homogeneous assay using AlphaScreen and AF.2A1. AF.2A1 is a 25-residue artificial protein that binds specifically to non-native IgG generated under chemical and physical stresses. This assay is performed in a short period of time. Our results show that AF.2A1-AlphaScreen may be used to evaluate the various types of IgG, as AF.2A1 recognizes the non-native structure in the constant region (Fc region) of IgG. The assay was effective for detection of non-native IgG, with particle size up to ca. 500 nm, generated under acid, heat, and stirring conditions. In addition, this technique is suitable for analyzing non-native IgG in CHO cell culture supernatant and mixed with large amounts of native IgG. These results indicate the potential of AF.2A1-AlphaScreen to be used as a high-throughput evaluation method for process monitoring as well as quality testing in the manufacturing of therapeutic IgG.
Collapse
Affiliation(s)
- Yukako Senga
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| | - Hiroshi Imamura
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| | - Takamitsu Miyafusa
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| | - Hideki Watanabe
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| | - Shinya Honda
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan.
| |
Collapse
|
30
|
On-chip immune cell activation and subsequent time-resolved magnetic bead-based cytokine detection. Biomed Microdevices 2017; 18:93. [PMID: 27628061 DOI: 10.1007/s10544-016-0117-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Cytokine profiling and immunophenotyping offer great potential for understanding many disease mechanisms, personalized diagnosis, and immunotherapy. Here, we demonstrate a time-resolved detection of cytokine from a single cell cluster using an in situ magnetic immune assay. An array of triple-layered microfluidic chambers was fabricated to enable simultaneous cell culture under perfusion flow and detection of the induced cytokines at multiple time-points. Each culture chamber comprises three fluidic compartments which are dedicated to, cell culture, perfusion and immunoassay. The three compartments are separated by porous membranes, which allow the diffusion of fresh nutrient from the perfusion compartment into the cell culture compartment and cytokines secretion from the cell culture compartment into the immune assay compartment. This structure hence enables capturing the released cytokines without disturbing the cell culture and without minimizing benefit gain from perfusion. Functionalized magnetic beads were used as a solid phase carrier for cytokine capturing and quantification. The cytokines released from differential stimuli were quantified in situ in non-differentiated U937 monocytes and differentiated macrophages.
Collapse
|
31
|
Yu ZTF, Joseph JG, Liu SX, Cheung MK, Haffey PJ, Kurabayashi K, Fu J. Centrifugal microfluidics for sorting immune cells from whole blood. SENSORS AND ACTUATORS. B, CHEMICAL 2017; 245:1050-1061. [PMID: 28966475 PMCID: PMC5619665 DOI: 10.1016/j.snb.2017.01.113] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Sorting and enumeration of immune cells from blood are critical operations involved in many clinical applications. Conventional methods for sorting and counting immune cells from blood, such as flow cytometry and hemocytometers, are tedious, inaccurate, and difficult for implementation for point-of-care (POC) testing. Herein we developed a microscale centrifugal technology termed Centrifugal Microfluidic Chip (CMC) capable of sorting immune cells from blood and in situ cellular analysis in a laboratory setting. Operation of the CMC entailed a blood specimen layered on a density gradient medium and centrifuged in microfluidic channels where immune cell subpopulations could rapidly be sorted into distinct layers according to their density differentials. We systematically studied effects of different blocking molecules for surface passivation of the CMC. We further demonstrated the applicability of CMCs for rapid separation of minimally processed human whole blood without affecting immune cell viability. Multi-color imaging and analysis of immune cell distributions and enrichment such as recovery and purity rates of peripheral blood mononuclear cells (PBMCs) were demonstrated using CMCs. Given its design and operation simplicity, portability, blood cell sorting efficiency, and in situ cellular analysis capability, the CMC holds promise for blood-based diagnosis and disease monitoring in POC applications.
Collapse
Affiliation(s)
- Zeta Tak For Yu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Jophin George Joseph
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Chemical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana, India 502285
| | - Shirley Xiaosu Liu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Mei Ki Cheung
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Parker James Haffey
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Katsuo Kurabayashi
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Michigan Center for Integrative Research in Critical Care, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
32
|
Tang CK, Vaze A, Rusling JF. Automated 3D-printed unibody immunoarray for chemiluminescence detection of cancer biomarker proteins. LAB ON A CHIP 2017; 17:484-489. [PMID: 28067370 PMCID: PMC5317057 DOI: 10.1039/c6lc01238h] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
A low cost three-dimensional (3D) printed clear plastic microfluidic device was fabricated for fast, low cost automated protein detection. The unibody device features three reagent reservoirs, an efficient 3D network for passive mixing, and an optically transparent detection chamber housing a glass capture antibody array for measuring chemiluminescence output with a CCD camera. Sandwich type assays were built onto the glass arrays using a multi-labeled detection antibody-polyHRP (HRP = horseradish peroxidase). Total assay time was ∼30 min in a complete automated assay employing a programmable syringe pump so that the protocol required minimal operator intervention. The device was used for multiplexed detection of prostate cancer biomarker proteins prostate specific antigen (PSA) and platelet factor 4 (PF-4). Detection limits of 0.5 pg mL-1 were achieved for these proteins in diluted serum with log dynamic ranges of four orders of magnitude. Good accuracy vs. ELISA was validated by analyzing human serum samples. This prototype device holds good promise for further development as a point-of-care cancer diagnostics tool.
Collapse
Affiliation(s)
- C K Tang
- Department of Chemistry, University of Connecticut, 55 North Eagleville Road, Storrs, Connecticut 06269-3060, USA.
| | - A Vaze
- Department of Chemistry, University of Connecticut, 55 North Eagleville Road, Storrs, Connecticut 06269-3060, USA.
| | - J F Rusling
- Department of Chemistry, University of Connecticut, 55 North Eagleville Road, Storrs, Connecticut 06269-3060, USA. and Institute of Materials Science, University of Connecticut, Storrs, Connecticut 06269, USA and Dept. of Surgery, and Neag Cancer Center, UConn Health, 263 Farmington Av., Farmington, Connecticut 06030, USA and School of Chemistry, National University of Ireland at Galway, Ireland
| |
Collapse
|
33
|
Nayak S, Blumenfeld NR, Laksanasopin T, Sia SK. Point-of-Care Diagnostics: Recent Developments in a Connected Age. Anal Chem 2017; 89:102-123. [PMID: 27958710 PMCID: PMC5793870 DOI: 10.1021/acs.analchem.6b04630] [Citation(s) in RCA: 294] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Samiksha Nayak
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA
| | - Nicole R. Blumenfeld
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA
| | - Tassaneewan Laksanasopin
- Biological Engineering Program, Faculty of Engineering, King Mongkut’s University of Technology Thonburi, 126 Pracha Uthit Rd., Bang Mod, Thung Khru, Bangkok 10140, Thailand
| | - Samuel K. Sia
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA
| |
Collapse
|
34
|
Moore CJ, Giovannini G, Kunc F, Hall AJ, Gubala V. ‘Overloading’ fluorescent silica nanoparticles with dyes to improve biosensor performance. J Mater Chem B 2017. [DOI: 10.1039/c7tb01284e] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Using dye-doped silica nanoparticles (DSNP) as reporter probes, we describe a simple method of enhancing fluorescent signal and the extension of the detectable target concentration range in a proof-of-concept ‘dissolution immunoassay’.
Collapse
Affiliation(s)
- Colin J. Moore
- Medway School of Pharmacy
- University of Kent
- Chatham Maritime
- Kent
- UK
| | | | - Filip Kunc
- Medway School of Pharmacy
- University of Kent
- Chatham Maritime
- Kent
- UK
| | - Andrew J. Hall
- Medway School of Pharmacy
- University of Kent
- Chatham Maritime
- Kent
- UK
| | - Vladimir Gubala
- Medway School of Pharmacy
- University of Kent
- Chatham Maritime
- Kent
- UK
| |
Collapse
|
35
|
Zhu L, Patel SH, Johnson M, Kale A, Raval Y, Tzeng TR, Xuan X. Enhanced Throughput for Electrokinetic Manipulation of Particles and Cells in a Stacked Microfluidic Device. MICROMACHINES 2016; 7:mi7090156. [PMID: 30404325 PMCID: PMC6190188 DOI: 10.3390/mi7090156] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 08/23/2016] [Accepted: 08/29/2016] [Indexed: 01/08/2023]
Abstract
Electrokinetic manipulation refers to the control of particle and cell motions using an electric field. It is an efficient technique for microfluidic applications with the ease of operation and integration. It, however, suffers from an intrinsic drawback of low throughput due to the linear dependence of the typically very low fluid permittivity. We demonstrate in this work a significantly enhanced throughput for electrokinetic manipulation of particles and cells by the use of multiple parallel microchannels in a two-layer stacked microfluidic device. The fabrication of this device is simple without the need of a precise alignment of the two layers. The number of layers and the number of microchannels in each layer can thus be further increased for a potentially high throughput electrokinetic particle and cell manipulations.
Collapse
Affiliation(s)
- Lin Zhu
- School of Engineering, Anhui Agricultural University, Hefei 230036, China.
| | - Saurin H Patel
- Department of Mechanical Engineering, Clemson University, Clemson, SC 29634-0921, USA.
| | - Mark Johnson
- Department of Mechanical Engineering, Clemson University, Clemson, SC 29634-0921, USA.
| | - Akshay Kale
- Department of Mechanical Engineering, Clemson University, Clemson, SC 29634-0921, USA.
| | - Yash Raval
- Department of Biological Sciences, Clemson University, Clemson, SC 29634-0314, USA.
| | - Tzuen-Rong Tzeng
- Department of Biological Sciences, Clemson University, Clemson, SC 29634-0314, USA.
| | - Xiangchun Xuan
- Department of Mechanical Engineering, Clemson University, Clemson, SC 29634-0921, USA.
| |
Collapse
|
36
|
Yu ZTF, Cheung MK, Liu SX, Fu J. Accelerated Biofluid Filling in Complex Microfluidic Networks by Vacuum-Pressure Accelerated Movement (V-PAM). SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:4521-30. [PMID: 27409528 PMCID: PMC6215695 DOI: 10.1002/smll.201601231] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 06/11/2016] [Indexed: 05/27/2023]
Abstract
Rapid fluid transport and exchange are critical operations involved in many microfluidic applications. However, conventional mechanisms used for driving fluid transport in microfluidics, such as micropumping and high pressure, can be inaccurate and difficult for implementation for integrated microfluidics containing control components and closed compartments. Here, a technology has been developed termed Vacuum-Pressure Accelerated Movement (V-PAM) capable of significantly enhancing biofluid transport in complex microfluidic environments containing dead-end channels and closed chambers. Operation of the V-PAM entails a pressurized fluid loading into microfluidic channels where gas confined inside can rapidly be dissipated through permeation through a thin, gas-permeable membrane sandwiched between microfluidic channels and a network of vacuum channels. Effects of different structural and operational parameters of the V-PAM for promoting fluid filling in microfluidic environments have been studied systematically. This work further demonstrates the applicability of V-PAM for rapid filling of temperature-sensitive hydrogels and unprocessed whole blood into complex irregular microfluidic networks such as microfluidic leaf venation patterns and blood circulatory systems. Together, the V-PAM technology provides a promising generic microfluidic tool for advanced fluid control and transport in integrated microfluidics for different microfluidic diagnosis, organs-on-chips, and biomimetic studies.
Collapse
Affiliation(s)
- Zeta Tak For Yu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Mei Ki Cheung
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Shirley Xiaosu Liu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Michigan Center for Integrative Research in Critical Care, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
37
|
Tang CK, Vaze A, Shen M, Rusling JF. High-Throughput Electrochemical Microfluidic Immunoarray for Multiplexed Detection of Cancer Biomarker Proteins. ACS Sens 2016; 1:1036-1043. [PMID: 27747294 DOI: 10.1021/acssensors.6b00256] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Microchip-based microfluidic electrochemical arrays hold great promise for fast, high-throughput multiplexed detection of cancer biomarker proteins at low cost per assay using relatively simple instrumentation. Here we describe an inexpensive high-throughput electrochemical array featuring 32 individually addressable microelectrodes that is further multiplexed with an 8-port manifold to provide 256 sensors. The gold electrode arrays were fabricated by wet-etching commercial gold compact discs (CD-R) followed by patterned insulation. A print-and-peel method was used to create sub-microliter hydrophobic wells surrounding each sensor to eliminate cross contamination during immobilization of capture antibodies. High-throughput analyses were realized using eight 32-sensor immunoarrays connected to the miniaturized 8-port manifold, allowing 256 measurements in <1 h. This system was used to determine prostate cancer biomarker proteins prostate specific antigen (PSA), prostate specific membrane antigen (PSMA), interleukin-6 (IL-6), and platelet factor-4 (PF-4) in serum. Clinically relevant detection limits (0.05 to 2 pg mL-1) and 5-decade dynamic ranges (sub pg mL-1 to well above ng mL-1) were achieved for these proteins utilizing precapture of analyte proteins on magnetic nanoparticles decorated with enzyme labels and antibodies.
Collapse
Affiliation(s)
| | | | | | - James F. Rusling
- Department
of Surgery and Neag Cancer Center, University of Connecticut Health Center, Farmington, Connecticut 06032, United States
- School
of Chemistry, National University of Ireland at Galway, Galway, Ireland
| |
Collapse
|
38
|
Dixit CK, Kadimisetty K, Otieno BA, Tang C, Malla S, Krause CE, Rusling JF. Electrochemistry-based approaches to low cost, high sensitivity, automated, multiplexed protein immunoassays for cancer diagnostics. Analyst 2016; 141:536-47. [PMID: 26525998 PMCID: PMC4701586 DOI: 10.1039/c5an01829c] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Early detection and reliable diagnostics are keys to effectively design cancer therapies with better prognoses. The simultaneous detection of panels of biomarker proteins holds great promise as a general tool for reliable cancer diagnostics. A major challenge in designing such a panel is to decide upon a coherent group of biomarkers which have higher specificity for a given type of cancer. The second big challenge is to develop test devices to measure these biomarkers quantitatively with high sensitivity and specificity, such that there are no interferences from the complex serum or tissue matrices. Lastly, integrating all these tests into a technology that does not require exclusive training to operate, and can be used at point-of-care (POC) is another potential bottleneck in futuristic cancer diagnostics. In this article, we review electrochemistry-based tools and technologies developed and/or used in our laboratories to construct low-cost microfluidic protein arrays for the highly sensitive detection of a panel of cancer-specific biomarkers with high specificity which at the same time has the potential to be translated into POC applications.
Collapse
Affiliation(s)
- Chandra K. Dixit
- Department of Chemistry, University of Connecticut, Storrs, CT 06269, USA
| | | | - Brunah A. Otieno
- Department of Chemistry, University of Connecticut, Storrs, CT 06269, USA
| | - Chi Tang
- Department of Chemistry, University of Connecticut, Storrs, CT 06269, USA
| | - Spundana Malla
- Department of Chemistry, University of Connecticut, Storrs, CT 06269, USA
| | - Colleen E. Krause
- Department of Chemistry, University of Hartford, West Hartford, CT 06117, USA
| | - James F. Rusling
- Department of Chemistry, University of Connecticut, Storrs, CT 06269, USA
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT 06269, USA
- School of Chemistry, National University of Ireland at Galway, Ireland
- Institute of Materials Science, University of Connecticut, Storrs, Connecticut 06269, United States
| |
Collapse
|