1
|
Hsu CY, Saleh RO, Mohammed JS, Mansuri N, Rekha MM, Kundlas M, Anand A, Sahoo S, Zwamel AH, Hulail HM. The dynamic interplay between melanoma cells and CAFs: Implications drug resistance and immune evasion and possible therapeutics. Exp Cell Res 2025; 449:114581. [PMID: 40311910 DOI: 10.1016/j.yexcr.2025.114581] [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: 03/13/2025] [Revised: 04/25/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
Melanoma, a malignancy of varying prognoses across primary sites (cutaneous, ocular, and mucosal), typically displays peculiar treatment challenges in metastatic and refractory settings. Cancer-associated fibroblasts (CAFs) have long been recognized as pivotal components within melanoma's tumor microenvironment (TME), originating from various sources and manifesting considerable heterogeneity. These cells actively produce extracellular matrix (ECM), induce angiogenesis, provide metabolic support, contribute to drug resistance, and feed tumor progression and metastasis. Among the many growth factors and cytokines they secrete, including TGF-β and IL-6, they aid in anti-tumor immunity by recruiting immunosuppressive cells and inhibiting cytotoxic T-cell activity, contributing to immune evasion. These dynamic cells sculpt the tumor's niche, allowing cancer cells to survive and proliferate, and their existence is widely correlated with poor prognosis. Taking a cue from the previously established groundwork of how the surroundings heavily influence tumor development, this review attempts to profile the intricate interaction of melanoma cells with the CAFs, the ECM, and signaling molecules. We explore different subtypes of CAFs, their origins, and how they have evolved in their pro-tumorigenic roles in melanoma. Additionally, we review recent advancements in the therapeutic arsenal targeting CAFs to achieve a more effective treatment response. By detailing the specific roles played by different CAFs subtypes in the modulation of immuno-responses and treatment outcomes, this review will further provide insights into the targeted therapy to disrupt CAFs-mediated tumor support in melanoma.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University, Tempe Campus, Phoenix, AZ, 85004, USA.
| | - Raed Obaid Saleh
- Department of Medical Laboratories Techniques, College of Health and Medical Techniques, University of Al Maarif, Al Anbar, 31001, Iraq.
| | - Jaafaru Sani Mohammed
- Medical Analysis Department, Faculty of Applied Science, Tishk International University, Erbil, Iraq
| | - Nasrin Mansuri
- Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Mayank Kundlas
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Alex Anand
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Samir Sahoo
- Department of General Medicine, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India
| | - Ahmed Hussein Zwamel
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University, Najaf, Iraq; Department of medical analysis, Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Department of medical analysis, Medical laboratory technique college, the Islamic University of Babylon, Babylon, Iraq
| | - Hanen Mahmod Hulail
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| |
Collapse
|
2
|
Jang W, Kim YJ, Roh HK, Song EL, Bong KW. Hydrogel-Based In Situ DNA Extension Assay for Multiplexed and Rapid Detection of MicroRNA. Anal Chem 2025; 97:7317-7324. [PMID: 40148761 DOI: 10.1021/acs.analchem.4c06995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
MicroRNAs (miRNAs) are important biomarkers for liquid biopsy, with extensive applicability to diverse diseases. Among diverse miRNA sensing platforms, graphically encoded hydrogel-based miRNA detection technology is a highly promising diagnostic tool, in terms of sensitivity, specificity, and multiplexing capability. However, the conventional hydrogel-based miRNA detection process suffers from a long assay time (more than 3 h) and redundant assay steps, limiting the practical applicability to actual clinical fields. In this study, we develop a hydrogel-based in situ DNA extension assay for rapid, simple, and multiplexed miRNA detection. Unlike typical hydrogel-based assays, the target hybridization and biotinylation for fluorophore labeling are integrated into a single step via target miRNA-primed DNA extension in hydrogel microparticles. Therefore, multiple microRNA targets can be quantitatively detected within 45 min by two assay steps composed of (1) target capture/biotinylation and (2) fluorophore labeling via streptavidin-biotin interaction. We validate robust sensitivities (down to the low picomolar level) and specificities (single-nucleotide level) by conducting singleplex assays for breast cancer-related miRNA markers (miR-16, miR-92a, and let-7a). Furthermore, multiplexed detection of these miRNA markers is conducted to validate robust multiplexing capacity with negligible nonspecific signal expression. Finally, multiple types of miRNAs in the lysate of breast cancer cells (MCF-7) are successfully detected using the developed assay. We expect the developed hydrogel-based assay can contribute to biomedical and omic fields, enabling high-throughput profiling of multiple miRNAs.
Collapse
Affiliation(s)
- Wookyoung Jang
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Yu Jin Kim
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Hyun Kyo Roh
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Republic of Korea
| | - E Loomee Song
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Ki Wan Bong
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Republic of Korea
| |
Collapse
|
3
|
Mukherjee A, Verma A, Das T, Ghosh B, Ghosh Z. Circulating microRNAs in Body Fluid: "Fingerprint" RNA Snippets Deeply Impact Reproductive Biology. Reprod Sci 2025; 32:555-574. [PMID: 39658771 DOI: 10.1007/s43032-024-01753-y] [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: 07/01/2024] [Accepted: 11/22/2024] [Indexed: 12/12/2024]
Abstract
Circulating miRNAs (C-miRNAs) occuring in a cell-free form within body fluids and other extracellular environments have garnered attention in recent times. They offer deeper insight into various physiological and pathological processes which include reproductive health. This review delves into their diagnostic potential across a spectrum of reproductive disorders, including conditions affecting ovarian function, male infertility and post pregnancy issues. Through analysis of C-miRNA profiles in bodily fluids, researchers uncover crucial markers indicative of reproductive challenges. Dysregulated C-miRNAs emerge as important players in the progression of several reproductive disorders which is the main focus of this review. Advancements in technology, facilitate precise detection and quantification of C-miRNAs, paving the way for innovative diagnostic approaches. Challenges in studying C-miRNAs, such as their low abundance and variability in expression levels, underscore the need for standardized protocols and rigorous validation methods. Despite these challenges, ongoing research endeavors aim to unravel the complex regulatory roles of C-miRNAs in reproductive biology, with potential implications for clinical practice and therapeutic interventions.
Collapse
Affiliation(s)
- Ayan Mukherjee
- Department of Animal Biotechnology, West Bengal University of Animal and Fishery Sciences, Mohanpur, West Bengal, 741252, India.
| | - Arpana Verma
- Department of Biological Sciences, Bose Institute, EN Block, Sector V, Kolkata, West Bengal, 700091, India
| | - Troyee Das
- Department of Biological Sciences, Bose Institute, EN Block, Sector V, Kolkata, West Bengal, 700091, India
| | - Byapti Ghosh
- Department of Biological Sciences, Bose Institute, EN Block, Sector V, Kolkata, West Bengal, 700091, India
| | - Zhumur Ghosh
- Department of Biological Sciences, Bose Institute, EN Block, Sector V, Kolkata, West Bengal, 700091, India.
| |
Collapse
|
4
|
Zhand S, Goss DM, Cheng YY, Warkiani ME. Recent Advances in Microfluidics for Nucleic Acid Analysis of Small Extracellular Vesicles in Cancer. Adv Healthc Mater 2025; 14:e2401295. [PMID: 39707658 DOI: 10.1002/adhm.202401295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 11/28/2024] [Indexed: 12/23/2024]
Abstract
Small extracellular vesicles (sEVs) are membranous vesicles released from cellular structures through plasma membrane budding. These vesicles contain cellular components such as proteins, lipids, mRNAs, microRNAs, long-noncoding RNA, circular RNA, and double-stranded DNA, originating from the cells they are shed from. Ranging in size from ≈25 to 300 nm and play critical roles in facilitating cell-to-cell communication by transporting signaling molecules. The discovery of sEVs in bodily fluids and their involvement in intercellular communication has revolutionized the fields of diagnosis, prognosis, and treatment, particularly in diseases like cancer. Conventional methods for isolating and analyzing sEVs, particularly their nucleic acid content face challenges including high costs, low purity, time-consuming processes, limited standardization, and inconsistent yield. The development of microfluidic devices, enables improved precision in sorting, isolating, and molecular-level separation using small sample volumes, and offers significant potential for the enhanced detection and monitoring of sEVs associated with cancer. These advanced techniques hold great promise for creating next-generation diagnostic and prognostic tools given their possibility of being cost-effective, simple to operate, etc. This comprehensive review explores the current state of research on microfluidic devices for the detection of sEV-derived nucleic acids as biomarkers and their translation into practical point-of-care and clinical applications.
Collapse
Affiliation(s)
- Sareh Zhand
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Dale Mark Goss
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Yuen Yee Cheng
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute of Molecular Theranostics, Sechenov First Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
5
|
Máčala J, Kuusinen S, Lahtinen S, Gorris HH, Skládal P, Farka Z, Soukka T. Amplification-Free Attomolar Detection of Short Nucleic Acids with Upconversion Luminescence: Eliminating Nonspecific Binding by Hybridization Complex Transfer. Anal Chem 2025; 97:1775-1782. [PMID: 39799476 PMCID: PMC11780573 DOI: 10.1021/acs.analchem.4c05401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/09/2024] [Accepted: 12/18/2024] [Indexed: 01/15/2025]
Abstract
The anti-Stokes emission of photon upconversion nanoparticles (UCNPs) facilitates their use as labels for ultrasensitive detection in biological samples as infrared excitation does not induce autofluorescence at visible wavelengths. The detection of extremely low-abundance analytes, however, remains challenging as it is impossible to completely avoid nonspecific binding of label conjugates. To overcome this limitation, we developed a novel hybridization complex transfer technique using UCNP labels to detect short nucleic acids directly without target amplification. The assay involves capturing the target-label complexes on an initial solid phase, then using releasing oligonucleotides to specifically elute only the target-UCNP complexes and recapturing them on another solid phase. The nonspecifically adsorbed labels remain on the first solid phase, enabling background-free, ultrasensitive detection. When magnetic microparticles were used as the first solid phase in a sample volume of 120 μL, the assay achieved a limit of detection (LOD) of 310 aM, a 27-fold improvement over the reference assay without transfer. Moreover, the additional target-specific steps introduced in the complex transfer procedure improved the sequence specificity of the complex transfer assay compared with the reference assay. The suitability for clinical analysis was confirmed using spiked plasma samples, resulting in an LOD of 190 aM. By increasing the sample volume to 600 μL and using magnetic preconcentration, the LOD was improved to 46 aM. These results highlight the importance of background elimination in achieving ultralow LODs for the analysis of low-abundance biomarkers.
Collapse
Affiliation(s)
- Jakub Máčala
- Department
of Biochemistry, Faculty of Science, Masaryk
University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Saara Kuusinen
- Department
of Life Technologies/Biotechnology, Faculty of Technology, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland
| | - Satu Lahtinen
- Department
of Life Technologies/Biotechnology, Faculty of Technology, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland
| | - Hans H. Gorris
- Department
of Biochemistry, Faculty of Science, Masaryk
University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Petr Skládal
- Department
of Biochemistry, Faculty of Science, Masaryk
University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Zdeněk Farka
- Department
of Biochemistry, Faculty of Science, Masaryk
University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Tero Soukka
- Department
of Life Technologies/Biotechnology, Faculty of Technology, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland
| |
Collapse
|
6
|
Shi Y, Wei J, Nie Y, Luo J, Chen T, Xi Q, Zhang Y, Sun J. Plant-derived miR166a-3p packaged into exosomes to cross-kingdom inhibit mammary cell proliferation and promote apoptosis by targeting APLNR gene. Int J Biol Macromol 2025; 286:138470. [PMID: 39645121 DOI: 10.1016/j.ijbiomac.2024.138470] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
Plant-derived microRNAs (miRNAs) have attracted significant attention for their potential in cross-kingdom gene regulation, but the mechanisms of their entry, stability, and function in animal bodies need further investigation. We provided an in-depth analysis of tissue-specific miRNA expression in dairy cows, identifying 347 miRNAs, including 16 novel candidates, across 21 normal tissues. Our findings revealed that specific miRNAs, such as miR-192, miR-143, miR-148a, miR-486, and miR-21-5p, showed distinct tissue enrichment. In addition, a total of 167 maize-derived miRNAs were identified in dairy cow tissues, particularly in the rumen, mammary glands, serum, and exosomes. These exogenous miRNAs, which are abundant and conserved among plants, may be absorbed by the SLC46A2 transporter in the rumen epithelium during feeding and distributed to other tissues via exosomal encapsulation. The maize-derived miR166a-3p was highly abundant. Transfection experiments confirmed that miR166a-3p reduces the expression of proliferation markers (PCNA, Cyclin D, and Cyclin E) and the anti-apoptotic gene Bcl2, while upregulating the pro-apoptotic gene Bax. Moreover, exosomes derived from bovine serum were found to mediate these effects, as miR166a-3p-enriched exosomes inhibited cell proliferation and promoted apoptosis, further supporting the cross-kingdom role of plant-derived miRNAs in regulating biological processes. This study enhances the understanding of miRNA regulatory mechanisms, particularly the absorption and systemic transport of plant-derived miRNAs in dairy cows. The findings underscore the potential for using exogenous miRNAs, like miR166a-3p, in agricultural and medical contexts, warranting further investigation into their functions and cross-species interactions.
Collapse
Affiliation(s)
- Yiru Shi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Junjie Wei
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Ying Nie
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Junyi Luo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Ting Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Qianyun Xi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Jiajie Sun
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.
| |
Collapse
|
7
|
Wang K, Wang S, Margolis S, Cho JM, Zhu E, Dupuy A, Yin J, Park SK, Magyar CE, Adeyiga OB, Jensen KS, Belperio JA, Passam F, Zhao P, Hsiai TK. Rapid prediction of acute thrombosis via nanoengineered immunosensors with unsupervised clustering for multiple circulating biomarkers. SCIENCE ADVANCES 2024; 10:eadq6778. [PMID: 39661669 PMCID: PMC11633740 DOI: 10.1126/sciadv.adq6778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 11/04/2024] [Indexed: 12/13/2024]
Abstract
The recent SARS-CoV-2 pandemic underscores the need for rapid and accurate prediction of clinical thrombotic events. Here, we developed nanoengineered multichannel immunosensors for rapid detection of circulating biomarkers associated with thrombosis, including C-reactive protein (CRP), calprotectin, soluble platelet selectin (sP-selectin), and D-dimer. We fabricated the immunosensors using fiber laser engraving of carbon nanotubes and CO2 laser cutting of microfluidic channels, along with the electrochemical deposition of gold nanoparticles to conjugate with biomarker-specific aptamers and antibody. Using unsupervised clustering based on four biomarker concentrations, we predicted thrombotic events in 49 of 53 patients. The four-biomarker combination yielded an area under the receiver operating characteristic curve (AUC) of 0.95, demonstrating high sensitivity and specificity for acute thrombosis prediction compared to the AUC values for individual biomarkers: CRP (0.773), calprotectin (0.711), sP-selectin (0.683), and D-dimer (0.739). Thus, a nanoengineered multichannel platform with unsupervised clustering provides accurate and efficient methods for predicting thrombosis, guiding personalized medicine.
Collapse
Affiliation(s)
- Kaidong Wang
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, CA 90095, USA
- Division of Cardiology, Department of Medicine, Greater Los Angeles VA Healthcare System, Los Angeles, CA 90073, USA
| | - Shaolei Wang
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Samuel Margolis
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Jae Min Cho
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Division of Cardiology, Department of Medicine, Greater Los Angeles VA Healthcare System, Los Angeles, CA 90073, USA
| | - Enbo Zhu
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Alexander Dupuy
- Department of Haematology, Royal Prince Alfred Hospital, Sydney, New South Wales 2050, Australia
- Central Clinical School, Faculty Medicine and Health, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Junyi Yin
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Seul-Ki Park
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Division of Cardiology, Department of Medicine, Greater Los Angeles VA Healthcare System, Los Angeles, CA 90073, USA
| | - Clara E. Magyar
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Oladunni B. Adeyiga
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Kristin Schwab Jensen
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - John A. Belperio
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Freda Passam
- Department of Haematology, Royal Prince Alfred Hospital, Sydney, New South Wales 2050, Australia
- Central Clinical School, Faculty Medicine and Health, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Peng Zhao
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Tzung K. Hsiai
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, CA 90095, USA
- Division of Cardiology, Department of Medicine, Greater Los Angeles VA Healthcare System, Los Angeles, CA 90073, USA
| |
Collapse
|
8
|
Devasinghe SU, Claus EL, Strait ME, Pagariya D, Anand RK. Electrokinetic Preconcentration and Label-Free Electrical Detection of SARS-CoV-2 RNA at a Packed Bed of Bioconjugated Microspheres. ACS Sens 2024; 9:5776-5781. [PMID: 39467261 DOI: 10.1021/acssensors.4c00427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
In this communication, we demonstrate the electrical detection of SARS-CoV-2 RNA at low femtomolar concentrations without labels or amplification reactions. Following its extraction from virus particles, the viral RNA was electrokinetically preconcentrated (100-fold) within a packed bed of probe-modified microbeads. This preconcentration was accomplished by counter-flow focusing of the RNA along an electric field gradient generated by faradaic ion concentration polarization (fICP). Hybridization of the 30 kb target RNA to the probe-modified beads sufficiently altered their surface charge to yield a measurable change in the ionic conductivity of the packed bed─a feature leveraged for electrical detection. When a single-stranded DNA probe was used, the sensitivity of this enrichment and sensing scheme was low picomolar. However, the utilization of an uncharged PNA probe improved the limit of detection to 3.4 × 106 viral copies/mL (22.5 fM SARS-CoV-2 RNA). These results are significant for three reasons. First, the sensitivity is remarkable, given the micrometer scale of both the beads and interstitial spaces. Additional gains in enrichment and sensitivity are anticipated as fundamental parametric studies and optimization are undertaken. Second, this study reveals the impact of the probe type on the sensitivity of microscale surface ion conduction (μSIC) sensors. Third, the RNA sensing approach has practical advantages including its utilization of off-the-shelf beads, a reagent-free approach, nonoptical readout, and low driving voltage, which render it amenable to point-of-care (POC) implementation.
Collapse
Affiliation(s)
- Sanduni U Devasinghe
- Department of Chemistry, Iowa State University, Ames, Iowa 50011-1021, United States
| | - Echo L Claus
- Department of Chemistry, Iowa State University, Ames, Iowa 50011-1021, United States
| | - Madison E Strait
- Department of Chemistry, Iowa State University, Ames, Iowa 50011-1021, United States
| | - Darshna Pagariya
- Department of Chemistry, Iowa State University, Ames, Iowa 50011-1021, United States
| | - Robbyn K Anand
- Department of Chemistry, Iowa State University, Ames, Iowa 50011-1021, United States
| |
Collapse
|
9
|
Wang T, Huang W, Gao X, Deng Y, Huang J. Single extracellular vesicle research: From cell population to a single cell. Biochem Biophys Res Commun 2024; 734:150439. [PMID: 39083971 DOI: 10.1016/j.bbrc.2024.150439] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024]
Abstract
Extracellular vesicles (EVs) are secreted by cells with a membrane structure and complex components such as DNA, RNA and proteins. These biomolecules play an important role in cell communication, cell proliferation, cell migration, vascularization, immune response and other physiological and pathological processes. Most current research on EVs focused on populations of EVs. Heterogeneity of EVs is neglected. Considering the heterogeneity of single EVs may offer critical molecular insights into cell-cell interactions, it is necessary to enhance our understanding about molecular characteristics from EVs derived from cell population to a single EV of derived from a single cell. This transformation is expected to provide a new insight into the understanding of cellular biology and the accurate description of the law of disease progress. In this article, we review the current research progress of single EV analysis technology for single EVs derived from cell population (SECP) and discuss its main applications in biological and clinical medicine research. After that, we propose the development direction, main difficulties and application prospect of single EV analysis technology for single EVs derived from single cells (SESC) according to our own research work, to provide new perspectives for the field of EV research.
Collapse
Affiliation(s)
- Ting Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wanqiu Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xu Gao
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yuliang Deng
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Jian Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
10
|
Ren X, Zhou R, Ronan G, Ozcebe SG, Ji J, Senapati S, March KL, Handberg E, Anderson D, Pepine CJ, Chang HC, Liu F, Zorlutuna P. Towards real-time myocardial infarction diagnosis: a convergence of machine learning and ion-exchange membrane technologies leveraging miRNA signatures. LAB ON A CHIP 2024; 24:5203-5214. [PMID: 39415669 PMCID: PMC11484500 DOI: 10.1039/d4lc00640b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024]
Abstract
Rapid diagnosis of acute myocardial infarction (AMI) is crucial for optimal patient management. Accurate diagnosis and time of onset of an acute event can influence treatment plans, such as percutaneous coronary intervention (PCI). PCI is most beneficial within 3 hours of AMI onset. MicroRNAs (miRNAs) are promising biomarkers, with potential of early AMI diagnosis, since they are released before cell death and subsequent release of larger molecules [e.g., cardiac troponins (cTn)], and have greater sensitivity and stability in plasma versus cTn regardless of timing of AMI onset. However, miRNA-based AMI diagnosis can result in false positives due to miRNA content overlap between AMI and stable coronary artery disease (CAD). Accordingly, we explored the possibility of using a miRNA profile, rather than a single miRNA, to distinguish between CAD and AMI, as well as different stages following AMI onset. First we screened a library of 800 miRNA using plasma samples from 4 patient cohorts; no known CAD, CAD, ST-segment elevation myocardial infarction (STEMI) and STEMI followed by PCI, using Nanostring miRNA profiling technology. From this screening, based on machine learning SCAD and Lasso algorithms, we identified 9 biomarkers (miR-200b, miR-543, miR-331, miR-3605, miR-301a, miR-18a, miR-423, miR-142, and miR-132) that were differentially expressed in CAD, STEMI and STEMI-PCI and explored them to identify a miRNA profile for rapid and accurate AMI diagnosis. These 9 miRNAs were selected as the most frequently identified targets by SCAD and Lasso, as indicated in the "drum-plot" model in the machine learning approach. We used age-matched patient samples to validate selected 9 miRNA biomarkers using a multiplexed ion-exchange membrane-based miRNA sensor platform, which measures specific miRNAs, and cTn as a control, simultaneously as a point-of-care device. Findings from this study will inform timely and accurate diagnosis of AMI and its stages, which are essential for effective management and optimal patient outcomes.
Collapse
Affiliation(s)
- Xiang Ren
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA.
| | - Ruyu Zhou
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN 46556, USA
| | - George Ronan
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA.
| | - S Gulberk Ozcebe
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA.
| | - Jiaying Ji
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA.
| | - Satyajyoti Senapati
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Keith L March
- Division of Cardiology, Department of Medicine in the College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Eileen Handberg
- Division of Cardiology, Department of Medicine in the College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - David Anderson
- Division of Cardiology, Department of Medicine in the College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Carl J Pepine
- Division of Cardiology, Department of Medicine in the College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Hsueh-Chia Chang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Fang Liu
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Pinar Zorlutuna
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA.
| |
Collapse
|
11
|
Song Y, Shi M, Wang Y. Deciphering the role of host-gut microbiota crosstalk via diverse sources of extracellular vesicles in colorectal cancer. Mol Med 2024; 30:200. [PMID: 39501131 PMCID: PMC11536884 DOI: 10.1186/s10020-024-00976-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 10/26/2024] [Indexed: 11/09/2024] Open
Abstract
Colorectal cancer is the most common type of cancer in the digestive system and poses a major threat to human health. The gut microbiota has been found to be a key factor influencing the development of colorectal cancer. Extracellular vesicles are important mediators of intercellular communication. Not only do they regulate life activities within the same individual, but they have also been found in recent years to be important mediators of communication between different species, such as the gut microbiota and the host. Their preventive, diagnostic, and therapeutic value in colorectal cancer is being explored. The aim of this review is to provide insights into the complex interactions between host and gut microbiota, particularly those mediated through extracellular vesicles, and how these interactions affect colorectal cancer development. In addition, the potential of extracellular vesicles from various body fluids as biomarkers was evaluated. Finally, we discuss the potential, challenges, and future research directions of extracellular vesicles in their application to colorectal cancer. Overall, extracellular vesicles have great potential for application in medical processes related to colorectal cancer, but their isolation and characterization techniques, intercellular communication mechanisms, and the effectiveness of their clinical application require further research and exploration.
Collapse
Affiliation(s)
- Yun Song
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai, 200336, PR China
| | - Min Shi
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai, 200336, PR China.
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, PR China.
| | - Yugang Wang
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai, 200336, PR China.
| |
Collapse
|
12
|
Dey B, Kundu S, Sundara BK. Polymeric Nanostructures Revolutionizing Cervical Cancer: Diagnostics, Therapeutics, and Theranostics. BIONANOSCIENCE 2024; 14:3906-3933. [DOI: 10.1007/s12668-024-01496-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2024] [Indexed: 01/05/2025]
Abstract
AbstractCervical cancer is the fourth most common cancer among women. Despite recent advancements in diagnostics and therapeutics, this disease is still a formidable challenge to deal with. Conventional methods for detecting human papillomavirus infection and imaging the tissues face major hurdles due to a lack of signal specificity and obscured resolution respectively. Moreover, chemotherapeutics struggle against the development of multidrug resistance and rapid clearance. With their easily tunable properties, polymeric nanostructures present a promising avenue for rapid, specific, and efficient diagnostics and therapeutics. These nanostructures also serve as theranostic agents that integrate imaging modalities with therapeutic approaches concurrently. This review highlights various types of polymeric nanostructures that serve as biosensors for the detection and quantification of cervical cancer biomarkers and act as nanocarriers for transporting fluorophores, photosensitizers, drugs, and radiosensitizers to their target site of action.
Graphical Abstract
Collapse
|
13
|
Goodrum R, Li H. Lysis of Extracellular Vesicles and Multiplexed Protein Detection via a Reverse Phase Immunoassay Using a Gold-Nanoparticle-Embedded Membrane Platform. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:22177-22189. [PMID: 39388120 DOI: 10.1021/acs.langmuir.4c02696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Extracellular vesicles (EVs) are cell-derived membrane-bound particles with molecular cargo reflective of their cell of origin. Analysis of disease-related EVs and associated cargo from biofluids is a promising tool for disease management. To facilitate the analysis of intravesicular molecules, EV lysis is needed. Moreover, highly sensitive and multiplexed detection methods are required to achieve early diagnostics. While cell lysis approaches have been well studied, the analysis of EV lysis methods and their effects on downstream molecular detection is lacking. In this work, we analyzed chemical, thermal, and mechanical EV lysis methods and determined their efficiency based on EV particle concentration and immunoassay activity. We, for the first time, discovered that vortex was an efficient EV lysis method and used it for detection of surface and intravesicular markers in a highly sensitive multiplexed reverse phase immunoassay on a gold-nanoparticle-embedded membrane. In phosphate-buffered saline, detection limits up to 3 orders of magnitude lower than enzyme-linked immunosorbent assay were achieved. In spiked human plasma, detection limits as low as 7.27 × 104 EVs/mL were achieved, making it suitable for early diagnostics. These results demonstrated an effective pipeline for lysing and molecular analysis of EVs from complex biofluids, paving the way for their broad applications in biomedicine.
Collapse
Affiliation(s)
- Rebecca Goodrum
- School of Engineering, University of Guelph, Guelph N1G2W1, Ontario, Canada
| | - Huiyan Li
- School of Engineering, University of Guelph, Guelph N1G2W1, Ontario, Canada
| |
Collapse
|
14
|
Jin Y, Wang J, Tang R, Jiang Y, Xi D. Nucleic Acid-Based Biological Nanopore Sensing Strategies for Tumor Marker Detection. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:21327-21340. [PMID: 39356337 DOI: 10.1021/acs.langmuir.4c02804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
Cancer, which is characterized by high mortality rates, poses a significant threat to global human health. Early diagnosis is of paramount importance in managing cancer, and tumor markers have emerged as crucial indicators for achieving this goal. The advent of precision medicine has further emphasized the need for the effective detection of these markers. However, traditional detection methods are hampered by numerous limitations. In recent years, nanopore technology has emerged as a promising alternative, due to its unique physical and chemical properties, which facilitate rapid, label-free, and amplification-free detection. This Review focuses on the direct detection of tumor markers through nucleic acid analysis and indirect detection mediated by nucleic acids and facilitated by biological nanopores. Furthermore, it also discusses the challenges and prospects of applying biological nanopore sensing technology in early cancer diagnosis, underscoring its potential to revolutionize tumor marker detection.
Collapse
Affiliation(s)
- Yameng Jin
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Chemistry and Chemical Engineering, Linyi University, Shandong 276005, China
| | - Junxiao Wang
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Chemistry and Chemical Engineering, Linyi University, Shandong 276005, China
| | - Ruping Tang
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Chemistry and Chemical Engineering, Linyi University, Shandong 276005, China
| | - Yao Jiang
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Life Science, Linyi University, Shandong 276005, China
| | - Dongmei Xi
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Life Science, Linyi University, Shandong 276005, China
| |
Collapse
|
15
|
Song J, Cho MH, Cho H, Song Y, Lee SW, Nam HC, Yoon TH, Shin JC, Hong JS, Kim Y, Ekanayake E, Jeon J, You DG, Im SG, Choi GS, Park JS, Carter BC, Balaj L, Seo AN, Miller MA, Park SY, Kang T, Castro CM, Lee H. Amplifying mutational profiling of extracellular vesicle mRNA with SCOPE. Nat Biotechnol 2024:10.1038/s41587-024-02426-6. [PMID: 39375445 DOI: 10.1038/s41587-024-02426-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 09/09/2024] [Indexed: 10/09/2024]
Abstract
Sequencing of messenger RNA (mRNA) found in extracellular vesicles (EVs) in liquid biopsies can provide clinical information such as somatic mutations, resistance profiles and tumor recurrence. Despite this, EV mRNA remains underused due to its low abundance in liquid biopsies, and large sample volumes or specialized techniques for analysis are required. Here we introduce Self-amplified and CRISPR-aided Operation to Profile EVs (SCOPE), a platform for EV mRNA detection. SCOPE leverages CRISPR-mediated recognition of target RNA using Cas13 to initiate replication and signal amplification, achieving a sub-attomolar detection limit while maintaining single-nucleotide resolution. As a proof of concept, we designed probes for key mutations in KRAS, BRAF, EGFR and IDH1 genes, optimized protocols for single-pot assays and implemented an automated device for multi-sample detection. We validated SCOPE's ability to detect early-stage lung cancer in animal models, monitored tumor mutational burden in patients with colorectal cancer and stratified patients with glioblastoma. SCOPE can expedite readouts, augmenting the clinical use of EVs in precision oncology.
Collapse
Affiliation(s)
- Jayeon Song
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Forensic Sciences, Sungkyunkwan University, Suwon, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Mi Hyeon Cho
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hayoung Cho
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Younseong Song
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | | | - Ho Chul Nam
- RevoSketch, Inc., Daejeon, Republic of Korea
| | - Tae Ho Yoon
- RevoSketch, Inc., Daejeon, Republic of Korea
| | | | - Jae-Sang Hong
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yejin Kim
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea
| | - Emil Ekanayake
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | - Jueun Jeon
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Dong Gil You
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | - Sung Gap Im
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Gyu-Seog Choi
- Colorectal Cancer Center, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jun Seok Park
- Colorectal Cancer Center, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Bob C Carter
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | - Leonora Balaj
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | - An Na Seo
- Department of Pathology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Miles A Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Soo Yeun Park
- Colorectal Cancer Center, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Taejoon Kang
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea.
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.
| | - Cesar M Castro
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA.
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Hakho Lee
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA.
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea.
| |
Collapse
|
16
|
Meng X, Wen K, Zhao J, Han Y, Ghandhi SA, Kaur SP, Brenner DJ, Turner HC, Amundson SA, Lin Q. Microfluidic measurement of intracellular mRNA with a molecular beacon probe towards point-of-care radiation triage. SENSORS & DIAGNOSTICS 2024; 3:1344-1352. [PMID: 39129862 PMCID: PMC11308381 DOI: 10.1039/d4sd00079j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 06/26/2024] [Indexed: 08/13/2024]
Abstract
In large-scale radiation exposure events, the ability to triage potential victims by the received radiation dosage is crucial. This can be evaluated by radiation-induced biological changes. Radiation-responsive mRNA is a class of biomarkers that has been explored for dose-dependency with methods such as RT-qPCR. However, these methods are challenging to implement for point-of-care devices. We have designed and used molecular beacons as probes for the measurement of radiation-induced changes of intracellular mRNA in a microfluidic device towards determining radiation dosage. Our experiments, in which fixed TK6 cells labeled with a molecular beacon specific to BAX mRNA exhibited dose-dependent fluorescence in a manner consistent with RT-qPCR analysis, demonstrate that such intracellular molecular probes can potentially be used in point-of-care radiation biodosimetry. This proof of concept could readily be extended to any RNA-based test to provide direct measurements at the bedside.
Collapse
Affiliation(s)
- Xin Meng
- Department of Mechanical Engineering, Columbia University New York NY 10027 USA
| | - Kechun Wen
- Department of Mechanical Engineering, Columbia University New York NY 10027 USA
| | - Jingyang Zhao
- Department of Mechanical Engineering, Columbia University New York NY 10027 USA
| | - Yaru Han
- Department of Mechanical Engineering, Columbia University New York NY 10027 USA
| | - Shanaz A Ghandhi
- Center for Radiological Research, Columbia University Irving Medical Center New York New York 10032 USA
| | - Salan P Kaur
- Center for Radiological Research, Columbia University Irving Medical Center New York New York 10032 USA
| | - David J Brenner
- Center for Radiological Research, Columbia University Irving Medical Center New York New York 10032 USA
| | - Helen C Turner
- Center for Radiological Research, Columbia University Irving Medical Center New York New York 10032 USA
| | - Sally A Amundson
- Center for Radiological Research, Columbia University Irving Medical Center New York New York 10032 USA
| | - Qiao Lin
- Department of Mechanical Engineering, Columbia University New York NY 10027 USA
| |
Collapse
|
17
|
Liu L, Cai J, Yang K, Sun B, Liu W, Li Y, Hu H. Molecular beacon-peptide probe based double recycling amplification for multiplexed detection of serum exosomal microRNAs. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:5202-5211. [PMID: 38994818 DOI: 10.1039/d4ay00629a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
Exosomal microRNAs (exomiRs) have been shown to play crucial roles as biomarkers for early detection and prognosis of cancer. However, simultaneous quantification of multiplex exomiRs is hindered by methods that require additional steps, such as labeling with fluorophores or gel visualization, which are susceptible to various factors. Herein, we developed a mass spectrometry-detectable and target-triggered method for multiplexed exomiR detection using three enzyme-based double recycling amplification in combination with well-designed molecular beacon-peptide (MBP) probes, called molecular beacon-peptide probe-based double recycling amplification (MBPDRA). MBP probes mediated the double recycling amplification reaction and were released as mass-detectable reporter peptides. In particular, the hybridization of the target microRNAs (miRNAs) with the stem-loop of the probe triggers two consecutive processes. The first cycle involved polymerase strand displacement amplification, leading to the production of complementary DNA (cycle I), and the second cycle encompassed the recycling exonuclease cleavage of the MBP probe (cycle II). Subsequently, excess probes were removed by interaction with streptavidin beads via biotin-streptavidin binding. The reporter peptides were released using trypsin and subsequently detected by mass spectrometry. Our method enables quantitative detection of multiple exomiRs with a dynamic range from 0.1 fM to 10 pM and a limit of quantification of 0.1 fM. Moreover, the proposed assay was successfully employed for quantification of three exomiRs, exmiR-21, exmiR-191, and exmiR-451a, in the sera of patients with pancreatic cancer. Based on these findings, we believe that the MBPDRA assay holds significant promise as a reliable method for quantifying multiple miRNAs in biomedical research and clinical diagnostics.
Collapse
Affiliation(s)
- Liang Liu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, 169 Donghu Road, Wuchang District, Wuhan 430071, China.
| | - Junlong Cai
- Department of Clinical Trial Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Kun Yang
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, 169 Donghu Road, Wuchang District, Wuhan 430071, China.
| | - Bo Sun
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, 222000, China
| | - Wei Liu
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, China
| | - Yang Li
- Department of Blood Transfusion, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuchang District, Wuhan, 430071, China.
| | - Hankun Hu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, 169 Donghu Road, Wuchang District, Wuhan 430071, China.
| |
Collapse
|
18
|
Kumar S, Senapati S, Chang HC. Extracellular vesicle and lipoprotein diagnostics (ExoLP-Dx) with membrane sensor: A robust microfluidic platform to overcome heterogeneity. BIOMICROFLUIDICS 2024; 18:041301. [PMID: 39056024 PMCID: PMC11272220 DOI: 10.1063/5.0218986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024]
Abstract
The physiological origins and functions of extracellular vesicles (EVs) and lipoproteins (LPs) propel advancements in precision medicine by offering non-invasive diagnostic and therapeutic prospects for cancers, cardiovascular, and neurodegenerative diseases. However, EV/LP diagnostics (ExoLP-Dx) face considerable challenges. Their intrinsic heterogeneity, spanning biogenesis pathways, surface protein composition, and concentration metrics complicate traditional diagnostic approaches. Commonly used methods such as nanoparticle tracking analysis, enzyme-linked immunosorbent assay, and nuclear magnetic resonance do not provide any information about their proteomic subfractions, including active proteins/enzymes involved in essential pathways/functions. Size constraints limit the efficacy of flow cytometry for small EVs and LPs, while ultracentrifugation isolation is hampered by co-elution with non-target entities. In this perspective, we propose a charge-based electrokinetic membrane sensor, with silica nanoparticle reporters providing salient features, that can overcome the interference, long incubation time, sensitivity, and normalization issues of ExoLP-Dx from raw plasma without needing sample pretreatment/isolation. A universal EV/LP standard curve is obtained despite their heterogeneities.
Collapse
Affiliation(s)
- Sonu Kumar
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Satyajyoti Senapati
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Hsueh-Chia Chang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| |
Collapse
|
19
|
Maniya NH, Kumar S, Franklin JL, Higginbotham JN, Scott AM, Gan HK, Coffey RJ, Senapati S, Chang HC. An anion exchange membrane sensor detects EGFR and its activity state in plasma CD63 extracellular vesicles from patients with glioblastoma. Commun Biol 2024; 7:677. [PMID: 38830977 PMCID: PMC11148014 DOI: 10.1038/s42003-024-06385-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 05/27/2024] [Indexed: 06/05/2024] Open
Abstract
We present a quantitative sandwich immunoassay for CD63 Extracellular Vesicles (EVs) and a constituent surface cargo, EGFR and its activity state, that provides a sensitive, selective, fluorophore-free and rapid alternative to current EV-based diagnostic methods. Our sensing design utilizes a charge-gating strategy, with a hydrophilic anion exchange membrane functionalized with capture antibodies and a charged silica nanoparticle reporter functionalized with detection antibodies. With sensitivity and robustness enhancement by the ion-depletion action of the membrane, this hydrophilic design with charged reporters minimizes interference from dispersed proteins, thus enabling direct plasma analysis without the need for EV isolation or sensor blocking. With a LOD of 30 EVs/μL and a high relative sensitivity of 0.01% for targeted proteomic subfractions, our assay enables accurate quantification of the EV marker, CD63, with colocalized EGFR by an operator/sample insensitive universal normalized calibration. We analysed untreated clinical samples of Glioblastoma to demonstrate this new platform. Notably, we target both total and "active" EGFR on EVs; with a monoclonal antibody mAb806 that recognizes a normally hidden epitope on overexpressed or mutant variant III EGFR. Analysis of samples yielded an area-under-the-curve (AUC) value of 0.99 and a low p-value of 0.000033, surpassing the performance of existing assays and markers.
Collapse
Affiliation(s)
- Nalin H Maniya
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Sonu Kumar
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Jeffrey L Franklin
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - James N Higginbotham
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Andrew M Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Hui K Gan
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Robert J Coffey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Satyajyoti Senapati
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA.
| | - Hsueh-Chia Chang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA.
| |
Collapse
|
20
|
Huang CK, Lin YN, Huang WS, Senapati S, Chang HC, Sun YM, Huang LF. RNA-based detection of genetically modified plants via current-voltage characteristic measurement. J Biotechnol 2024; 383:27-38. [PMID: 38336281 DOI: 10.1016/j.jbiotec.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/30/2024] [Accepted: 02/05/2024] [Indexed: 02/12/2024]
Abstract
The widespread adoption of genetically modified (GM) crops has escalated concerns about their safety and ethical implications, underscoring the need for efficient GM crop detection methods. Conventional detection methods, such as polymerase chain reaction, can be costly, lab-bound, and time-consuming. To overcome these challenges, we have developed RapiSense, a cost-effective, portable, and sensitive biosensor platform. This sensor generates a measurable voltage shift (0.1-1 V) in the system's current-voltage characteristics, triggered by an increase in membrane's negative charge upon hybridization of DNA/RNA targets with a specific DNA probe. Probes designed to identify the herbicide resistance gene hygromycin phosphotransferase show a detection range from ∼1 nM to ∼10 μM and can discriminate between complementary, non-specific, and mismatched nucleotide targets. The incorporation of a small membrane sensor to detect fragmented RNA samples substantially improve the platform's sensitivity. In this study, RapiSense has been effectively used to detect specific DNA and fragmented RNA in transgenic variants of Arabidopsis, sweet potato, and rice, showcasing its potential for rapid, on-site GM crop screening.
Collapse
Affiliation(s)
- Chun-Kai Huang
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan, Republic of China; Department of Chemical Engineering and Materials Science, Yuan Ze University, Taoyuan 320315, Taiwan, Republic of China; Institute of Plant and Microbial Biology, Academia Sinica, Taipei 115201, Taiwan, Republic of China
| | - Yi-Nan Lin
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan, Republic of China; Department of Chemical Engineering and Materials Science, Yuan Ze University, Taoyuan 320315, Taiwan, Republic of China
| | - Wen-Shan Huang
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan, Republic of China; Department of Chemical Engineering and Materials Science, Yuan Ze University, Taoyuan 320315, Taiwan, Republic of China
| | - Satyajyoti Senapati
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Hsueh-Chia Chang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Yi-Ming Sun
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Taoyuan 320315, Taiwan, Republic of China; R&D Center for Membrane Technology, Chung Yuan University, Taoyuan 320071, Taiwan, Republic of China
| | - Li-Fen Huang
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan, Republic of China.
| |
Collapse
|
21
|
Al-Hawary SIS, Saleh RO, Mansouri S, Noraldeen SAM, Alawadi AH, Kareem AH, Alsalamy A, Shakir MN, Alasheqi MQ, Mustafa YF. Isothermal amplification methods in cancer-related miRNA detection; a new paradigm in study of cancer pathology. Pathol Res Pract 2024; 254:155072. [PMID: 38228039 DOI: 10.1016/j.prp.2023.155072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 12/22/2023] [Accepted: 12/28/2023] [Indexed: 01/18/2024]
Abstract
MicroRNAs (miRNAs) are short, non-coding RNA molecules that regulate gene expression. They are involved in a wide range of biological processes, including development, differentiation, cell cycle regulation, and response to stress. Numerous studies have demonstrated that miRNAs are present in different bodily fluids, which could serve as an important biomarker. The advancement of techniques and strategies for the identification of cancer-associated miRNAs in human specimens offers a novel opportunity to diagnose cancer in early stages, predict patient prognosis and evaluate response to treatment. Isothermal techniques including loop-mediated isothermal amplification (LAMP), rolling circle amplification (RCA), or recombinase polymerase amplification (RPA) offer simplicity, efficiency, and rapidity in miRNA detection processes. In contrast to traditional PCR (polymerase chain reaction), these techniques analysis and quantify miRNA molecules in specimens using a single constant temperature. In this comprehensive review, we summarized the recent advances in cancer-related miRNA detection via highly sensitive isothermal amplification methods by more focusing on the involved mechanism.
Collapse
Affiliation(s)
| | - Raed Obaid Saleh
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Iraq
| | - Sofiene Mansouri
- Department of Biomedical Technology, College of Applied Medical Sciences in Al-Kharj, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; University of Tunis El Manar, Higher Institute of Medical Technologies of Tunis, Laboratory of Biophysics and Medical Technologies, Tunis, Tunisia.
| | | | - Ahmed Hussien Alawadi
- College of Technical Engineering, University of Najaf, Najaf, Iraq; College of Technical Engineering, the Islamic University of Al Diwaniyah, Iraq; College of Technical Engineering, the Islamic University of Babylon, Iraq
| | | | - Ali Alsalamy
- College of Technical Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna 66002, Iraq
| | - Maha Noori Shakir
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | | | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| |
Collapse
|
22
|
Giordano C, Accattatis FM, Gelsomino L, Del Console P, Győrffy B, Giuliano M, Veneziani BM, Arpino G, De Angelis C, De Placido P, Pietroluongo E, Zinno F, Bonofiglio D, Andò S, Barone I, Catalano S. miRNAs in the Box: Potential Diagnostic Role for Extracellular Vesicle-Packaged miRNA-27a and miRNA-128 in Breast Cancer. Int J Mol Sci 2023; 24:15695. [PMID: 37958677 PMCID: PMC10649351 DOI: 10.3390/ijms242115695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
Circulating extracellular vesicle (EV)-derived microRNAs (miRNAs) are now considered the next generation of cancer "theranostic" tools, with strong clinical relevance. Although their potential in breast cancer diagnosis has been widely reported, further studies are still required to address this challenging issue. The present study examined the expression profiles of EV-packaged miRNAs to identify novel miRNA signatures in breast cancer and verified their diagnostic accuracy. Circulating EVs were isolated from healthy controls and breast cancer patients and characterized following the MISEV 2018 guidelines. RNA-sequencing and real-time PCR showed that miRNA-27a and miRNA-128 were significantly down-regulated in patient-derived EVs compared to controls in screening and validation cohorts. Bioinformatics analyses of miRNA-target genes indicated several enriched biological processes/pathways related to breast cancer. Receiver operating characteristic (ROC) curves highlighted the ability of these EV-miRNAs to distinguish breast cancer patients from non-cancer controls. According to other reports, the levels of EV-miRNA-27a and EV-miRNA-128 are not associated with their circulating ones. Finally, evidence from the studies included in our systematic review underscores how the expression of these miRNAs in biofluids is still underinvestigated. Our findings unraveled the role of serum EV-derived miRNA-27a and miRNA-128 in breast cancer, encouraging further investigation of these two miRNAs within EVs towards improved breast cancer detection.
Collapse
Affiliation(s)
- Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy; (F.M.A.); (L.G.); (P.D.C.); (D.B.); (S.A.); (I.B.)
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
- Clinical Laboratory Unit, A.O. “Annunziata”, 87100 Cosenza, Italy
| | - Felice Maria Accattatis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy; (F.M.A.); (L.G.); (P.D.C.); (D.B.); (S.A.); (I.B.)
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Luca Gelsomino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy; (F.M.A.); (L.G.); (P.D.C.); (D.B.); (S.A.); (I.B.)
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Piercarlo Del Console
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy; (F.M.A.); (L.G.); (P.D.C.); (D.B.); (S.A.); (I.B.)
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Balázs Győrffy
- Departments of Bioinformatics and Pediatrics, Semmelweis University, 1094 Budapest, Hungary;
- TTK Cancer Biomarker Research Group, 1117 Budapest, Hungary
| | - Mario Giuliano
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80133 Naples, Italy; (M.G.); (G.A.); (C.D.A.); (P.D.P.); (E.P.)
| | - Bianca Maria Veneziani
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80133 Naples, Italy;
| | - Grazia Arpino
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80133 Naples, Italy; (M.G.); (G.A.); (C.D.A.); (P.D.P.); (E.P.)
| | - Carmine De Angelis
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80133 Naples, Italy; (M.G.); (G.A.); (C.D.A.); (P.D.P.); (E.P.)
| | - Pietro De Placido
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80133 Naples, Italy; (M.G.); (G.A.); (C.D.A.); (P.D.P.); (E.P.)
| | - Erica Pietroluongo
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80133 Naples, Italy; (M.G.); (G.A.); (C.D.A.); (P.D.P.); (E.P.)
| | - Francesco Zinno
- Immunohaematology and Transfusion Medicine, A.O. “Annunziata”, 87100 Cosenza, Italy;
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy; (F.M.A.); (L.G.); (P.D.C.); (D.B.); (S.A.); (I.B.)
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy; (F.M.A.); (L.G.); (P.D.C.); (D.B.); (S.A.); (I.B.)
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy; (F.M.A.); (L.G.); (P.D.C.); (D.B.); (S.A.); (I.B.)
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy; (F.M.A.); (L.G.); (P.D.C.); (D.B.); (S.A.); (I.B.)
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata di Rende (CS), 87036 Cosenza, Italy
- Clinical Laboratory Unit, A.O. “Annunziata”, 87100 Cosenza, Italy
| |
Collapse
|
23
|
Maniya NH, Kumar S, Franklin JL, Higginbotham JN, Scott AM, Gan HK, Coffey RJ, Senapati S, Chang HC. Detection of EGFR and its Activity State in Plasma CD63-EVs from Glioblastoma Patients: Rapid Profiling using an Anion Exchange Membrane Sensor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.16.562628. [PMID: 37905113 PMCID: PMC10614888 DOI: 10.1101/2023.10.16.562628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
We present a novel quantitative immunoassay for CD63 EVs (extracellular vesicles) and a constituent surface cargo, EGFR and its activity state, that provides a sensitive, selective, fluorophore-free and rapid alternative to current EV-based diagnostic methods. Our sensing design utilizes a charge-gating strategy, with a hydrophilic anion exchange membrane and a charged silica nanoparticle reporter. With sensitivity and robustness enhancement by the ion-depletion action of the membrane, this hydrophilic design with charged reporters minimizes interference from dispersed proteins and fluorophore degradation, thus enabling direct plasma analysis. With a limit of detection of 30 EVs/μL and a high relative sensitivity of 0.01% for targeted proteomic subfractions, our assay enables accurate quantification of the EV marker, CD63, with colocalized EGFR by an operator/sample insensitive universal normalized calibration. Glioblastoma necessitates improved non-invasive diagnostic approaches for early detection and monitoring. Notably, we target both total and "active" EGFR on EVs; with a monoclonal antibody mAb806 that recognizes a normally hidden epitope on overexpressed or mutant variant III EGFR. This approach offers direct glioblastoma detection from untreated human patient samples. Analysis of glioblastoma clinical samples yielded an area-under-the-curve (AUC) value of 0.99 and low p-value of 0.000033, significantly surpassing the performance of existing assays and markers.
Collapse
|
24
|
Cha B, Lee SH, Iqrar SA, Yi HG, Kim J, Park J. Rapid acoustofluidic mixing by ultrasonic surface acoustic wave-induced acoustic streaming flow. ULTRASONICS SONOCHEMISTRY 2023; 99:106575. [PMID: 37683414 PMCID: PMC10495656 DOI: 10.1016/j.ultsonch.2023.106575] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 08/18/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023]
Abstract
Ultrasonic surface acoustic wave (SAW)-induced acoustic streaming flow (ASF) has been utilized for microfluidic flow control, patterning, and mixing. Most previous research employed cross-type SAW acousto-microfluidic mixers, in which the SAWs propagated perpendicular to the flow direction. In this configuration, the flow mixing was induced predominantly by the horizontal component of the acoustic force, which was usually much smaller than the vertical component, leading to energy inefficiency and limited controllability. Here, we propose a vertical-type ultrasonic SAW acousto-microfluidic mixer to achieve rapid flow mixing with improved efficiency and controllability. We conducted in-depth numerical and experimental investigations of the vertical-type SAW-induced ASF to elucidate the acousto-hydrodynamic phenomenon under varying conditions of total flow rate, acoustic wave amplitude, and fluid viscosity conditions. We conducted computational fluid dynamics simulations for numerical flow visualization and utilized micro-prism-embedded microchannels for experimental flow visualization for the vertical SAW-induced ASF. We found that the SAW-induced vortices served as a hydrodynamic barrier for the co-flow streams for controlled flow mixing in the proposed device. For proof-of-concept application, we performed chemical additive-free rapid red blood cell lysis and achieved rapid cell lysis with high lysis efficiency based on the physical interactions of the suspended cells with the SAW-induced acoustic vortical flows. We believe that the proposed vertical-type ultrasonic SAW-based mixer can be broadly utilized for various microfluidic applications that require rapid, controlled flow mixing.
Collapse
Affiliation(s)
- Beomseok Cha
- Department of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Song Ha Lee
- Department of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Syed Atif Iqrar
- College of Engineering and Physical Sciences, Aston Institute of Photonic Technologies, Aston University, Birmingham B4 7ET, United Kingdom
| | - Hee-Gyeong Yi
- Department of Convergence Biosystems Engineering, College of Agriculture and Life Sciences, Chonnam National University, Gwangju 61186, Republic of Korea; Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jangho Kim
- Department of Convergence Biosystems Engineering, College of Agriculture and Life Sciences, Chonnam National University, Gwangju 61186, Republic of Korea; Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jinsoo Park
- Department of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
25
|
Juska VB, Maxwell G, Estrela P, Pemble ME, O'Riordan A. Silicon microfabrication technologies for biology integrated advance devices and interfaces. Biosens Bioelectron 2023; 237:115503. [PMID: 37481868 DOI: 10.1016/j.bios.2023.115503] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 06/25/2023] [Accepted: 06/26/2023] [Indexed: 07/25/2023]
Abstract
Miniaturization is the trend to manufacture ever smaller devices and this process requires knowledge, experience, understanding of materials, manufacturing techniques and scaling laws. The fabrication techniques used in semiconductor industry deliver an exceptionally high yield of devices and provide a well-established platform. Today, these miniaturized devices are manufactured with high reproducibility, design flexibility, scalability and multiplexed features to be used in several applications including micro-, nano-fluidics, implantable chips, diagnostics/biosensors and neural probes. We here provide a review on the microfabricated devices used for biology driven science. We will describe the ubiquity of the use of micro-nanofabrication techniques in biology and biotechnology through the fabrication of high-aspect-ratio devices for cell sensing applications, intracellular devices, probes developed for neuroscience-neurotechnology and biosensing of the certain biomarkers. Recently, the research on micro and nanodevices for biology has been progressing rapidly. While the understanding of the unknown biological fields -such as human brain- has been requiring more research with advanced materials and devices, the development protocols of desired devices has been advancing in parallel, which finally meets with some of the requirements of biological sciences. This is a very exciting field and we aim to highlight the impact of micro-nanotechnologies that can shed light on complex biological questions and needs.
Collapse
Affiliation(s)
- Vuslat B Juska
- Tyndall National Institute, University College Cork, T12R5CP, Ireland.
| | - Graeme Maxwell
- Tyndall National Institute, University College Cork, T12R5CP, Ireland
| | - Pedro Estrela
- Department of Electronic and Electrical Engineering, University of Bath, Bath, BA2 7AY, United Kingdom; Centre for Bioengineering & Biomedical Technologies (CBio), University of Bath, Bath, BA2 7AY, United Kingdom
| | | | - Alan O'Riordan
- Tyndall National Institute, University College Cork, T12R5CP, Ireland
| |
Collapse
|
26
|
Kim HJ, Rames MJ, Goncalves F, Kirschbaum CW, Roskams-Hieter B, Spiliotopoulos E, Briand J, Doe A, Estabrook J, Wagner JT, Demir E, Mills G, Ngo TTM. Selective enrichment of plasma cell-free messenger RNA in cancer-associated extracellular vesicles. Commun Biol 2023; 6:885. [PMID: 37644220 PMCID: PMC10465482 DOI: 10.1038/s42003-023-05232-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 08/09/2023] [Indexed: 08/31/2023] Open
Abstract
Extracellular vesicles (EVs) have been shown as key mediators of extracellular small RNA transport. However, carriers of cell-free messenger RNA (cf-mRNA) in human biofluids and their association with cancer remain poorly understood. Here, we performed a transcriptomic analysis of size-fractionated plasma from lung cancer, liver cancer, multiple myeloma, and healthy donors. Morphology and size distribution analysis showed the successful separation of large and medium particles from other soluble plasma protein fractions. We developed a strategy to purify and sequence ultra-low amounts of cf-mRNA from particle and protein enriched subpopulations with the implementation of RNA spike-ins to control for technical variability and to normalize for intrinsic drastic differences in cf-mRNA amount carried in each plasma fraction. We found that the majority of cf-mRNA was enriched and protected in EVs with remarkable stability in RNase-rich environments. We observed specific enrichment patterns of cancer-associated cf-mRNA in each particle and protein enriched subpopulation. The EV-enriched differentiating genes were associated with specific biological pathways, such as immune systems, liver function, and toxic substance regulation in lung cancer, liver cancer, and multiple myeloma, respectively. Our results suggest that dissecting the complexity of EV subpopulations illuminates their biological significance and offers a promising liquid biopsy approach.
Collapse
Affiliation(s)
- Hyun Ji Kim
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA
| | - Matthew J Rames
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA
| | - Florian Goncalves
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA
| | - C Ward Kirschbaum
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Breeshey Roskams-Hieter
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Elias Spiliotopoulos
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Josephine Briand
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Aaron Doe
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Joseph Estabrook
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
- Computational Biology Program, Oregon Health and Science University, Portland, OR, USA
| | - Josiah T Wagner
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
- Molecular Genomics Laboratory, Providence Health and Services, Portland, OR, USA
| | - Emek Demir
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
- Computational Biology Program, Oregon Health and Science University, Portland, OR, USA
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Gordon Mills
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Thuy T M Ngo
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA.
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA.
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA.
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
27
|
Rahn KL, Peramune U, Zhang T, Anand RK. Label-Free Electrochemical Methods for Disease Detection. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2023; 16:49-69. [PMID: 36854209 DOI: 10.1146/annurev-anchem-091622-085754] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Label-free electrochemical biosensing leverages the advantages of label-free techniques, low cost, and fewer user steps, with the sensitivity and portability of electrochemical analysis. In this review, we identify four label-free electrochemical biosensing mechanisms: (a) blocking the electrode surface, (b) allowing greater access to the electrode surface, (c) changing the intercalation or electrostatic affinity of a redox probe to a biorecognition unit, and (d) modulating ion or electron transport properties due to conformational and surface charge changes. Each mechanism is described, recent advancements are summarized, and relative advantages and disadvantages of the techniques are discussed. Furthermore, two avenues for gaining further diagnostic information from label-free electrochemical biosensors, through multiplex analysis and incorporating machine learning, are examined.
Collapse
Affiliation(s)
- Kira L Rahn
- Department of Chemistry, Colorado State University, Fort Collins, Colorado, USA
- Department of Chemistry, Iowa State University, Ames, Iowa, USA;
| | - Umesha Peramune
- Department of Chemistry, Iowa State University, Ames, Iowa, USA;
| | - Tianyi Zhang
- Department of Chemistry, Iowa State University, Ames, Iowa, USA;
| | - Robbyn K Anand
- Department of Chemistry, Iowa State University, Ames, Iowa, USA;
| |
Collapse
|
28
|
Ganchenko GS, Alekseev MS, Moroz IA, Mareev SA, Shelistov VS, Demekhin EA. Electrokinetic and Electroconvective Effects in Ternary Electrolyte Near Ion-Selective Microsphere. MEMBRANES 2023; 13:membranes13050503. [PMID: 37233564 DOI: 10.3390/membranes13050503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 05/27/2023]
Abstract
The paper presents theoretical and experimental investigations of the behavior of an electrolyte solution with three types of ions near an ion-selective microparticle with electrokinetically and pressure-driven flow. A special experimental cell has been developed for the investigations. An anion-selective spherical particle composed of ion-exchange resin is fixed in the center of the cell. An enriched region with a high salt concentration appears at the anode side of the particle when an electric field is turned on, according to the nonequilibrium electrosmosis behavior. A similar region exists near a flat anion-selective membrane. However, the enriched region near the particle produces a concentration jet that spreads downstream akin to a wake behind an axisymmetrical body. The fluorescent cations of Rhodamine-6G dye are chosen as the third species in the experiments. The ions of Rhodamine-6G have a 10-fold lower diffusion coefficient than the ions of potassium while bearing the same valency. This paper shows that the concentration jet behavior is described accurately enough with the mathematical model of a far axisymmetric wake behind a body in a fluid flow. The third species also forms an enriched jet, but its distribution turns out to be more complex. The concentration of the third species increases in the jet with an increase in pressure gradient. The pressure-driven flow stabilizes the jet, yet electroconvection has been observed near the microparticle for sufficiently strong electric fields. The electrokinetic instability and the electroconvection partially destroy the concentration jet of salt and the third species. The conducted experiments show good qualitative agreement with the numerical simulations. The presented results could be used in future for implementing microdevices based on membrane technology for solving problems of detection and preconcentration, and thus simplifying chemical and medical analyses utilizing the superconcentration phenomenon. Such devices are called membrane sensors, and are actively being studied.
Collapse
Affiliation(s)
- Georgy S Ganchenko
- Laboratory of Micro- and Nanoscale Electro- and Hydrodynamics, Financial University under the Government of the Russian Federation, 53 Leningradsky Prospect str., Moscow 125167, Russia
| | - Maxim S Alekseev
- Laboratory of Micro- and Nanoscale Electro- and Hydrodynamics, Financial University under the Government of the Russian Federation, 53 Leningradsky Prospect str., Moscow 125167, Russia
- Membrane Institute, Kuban State University, 149 Stavropolskaya str., Krasnodar 350040, Russia
| | - Ilya A Moroz
- Membrane Institute, Kuban State University, 149 Stavropolskaya str., Krasnodar 350040, Russia
| | - Semyon A Mareev
- Membrane Institute, Kuban State University, 149 Stavropolskaya str., Krasnodar 350040, Russia
| | - Vladimir S Shelistov
- Laboratory of Micro- and Nanoscale Electro- and Hydrodynamics, Financial University under the Government of the Russian Federation, 53 Leningradsky Prospect str., Moscow 125167, Russia
| | - Evgeny A Demekhin
- Laboratory of Micro- and Nanoscale Electro- and Hydrodynamics, Financial University under the Government of the Russian Federation, 53 Leningradsky Prospect str., Moscow 125167, Russia
- Laboratory of General Aeromechanics, Institute of Mechanics, Moscow State University, 1 Michurinsky Prospect, Moscow 119192, Russia
| |
Collapse
|
29
|
Hsu CC, Yang Y, Kannisto E, Zeng X, Yu G, Patnaik SK, Dy GK, Reid ME, Gan Q, Wu Y. Simultaneous Detection of Tumor Derived Exosomal Protein-MicroRNA Pairs with an Exo-PROS Biosensor for Cancer Diagnosis. ACS NANO 2023; 17:8108-8122. [PMID: 37129374 DOI: 10.1021/acsnano.2c10970] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Tumor derived exosomes (TEXs) have emerged as promising biomarkers for cancer liquid biopsy. Conventional methods (such as ELISA and qRT-PCR) and emerging biosensing technologies mainly detect a single type of exosomal biomarker due to the distinct properties of different biomolecules. Sensitive detection of two different types of TEX biomarkers, i.e., protein and microRNA combined biomarkers, may greatly improve cancer diagnostic accuracy. We developed an exosome protein microRNA one-stop (Exo-PROS) biosensor that not only selectively captured TEXs but also enabled in situ, simultaneous detection of TEX protein-microRNA pairs via a surface plasmon resonance mechanism. Exo-PROS assay is a fast, reliable, low sample consumption, and user-friendly test. With a total of 175 cancer patients and normal controls, we demonstrated that TEX protein-microRNA pairs measured by Exo-PROS assay detected lung cancer and breast cancer with 99% and 96% accuracy, respectively. Exo-PROS assay also showed superior diagnostic performance to conventional ELISA and qRT-PCR methods. Our results demonstrated that Exo-PROS assay is a potent liquid biopsy assay for cancer diagnosis.
Collapse
Affiliation(s)
- Chang-Chieh Hsu
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| | - Yunchen Yang
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| | - Eric Kannisto
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, New York 14263, United States
| | - Xie Zeng
- Department of Electrical Engineering, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| | - Guan Yu
- Department of Biostatistics, University at Buffalo, The State University of New York, Buffalo, New York 14263, United States
| | - Santosh K Patnaik
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, New York 14263, United States
| | - Grace K Dy
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, New York 14263, United States
| | - Mary E Reid
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, New York 14263, United States
| | - Qiaoqiang Gan
- Department of Electrical Engineering, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
- Materials Science Engineering, King Abdullah University of Science and Technology, Thuwal 23955-6900, Saudi Arabia
| | - Yun Wu
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| |
Collapse
|
30
|
Rey Gomez LM, Hirani R, Care A, Inglis DW, Wang Y. Emerging Microfluidic Devices for Sample Preparation of Undiluted Whole Blood to Enable the Detection of Biomarkers. ACS Sens 2023; 8:1404-1421. [PMID: 37011238 DOI: 10.1021/acssensors.2c02696] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
Blood testing allows for diagnosis and monitoring of numerous conditions and illnesses; it forms an essential pillar of the health industry that continues to grow in market value. Due to the complex physical and biological nature of blood, samples must be carefully collected and prepared to obtain accurate and reliable analysis results with minimal background signal. Examples of common sample preparation steps include dilutions, plasma separation, cell lysis, and nucleic acid extraction and isolation, which are time-consuming and can introduce risks of sample cross-contamination or pathogen exposure to laboratory staff. Moreover, the reagents and equipment needed can be costly and difficult to obtain in point-of-care or resource-limited settings. Microfluidic devices can perform sample preparation steps in a simpler, faster, and more affordable manner. Devices can be carried to areas that are difficult to access or that do not have the resources necessary. Although many microfluidic devices have been developed in the last 5 years, few were designed for the use of undiluted whole blood as a starting point, which eliminates the need for blood dilution and minimizes blood sample preparation. This review will first provide a short summary on blood properties and blood samples typically used for analysis, before delving into innovative advances in microfluidic devices over the last 5 years that address the hurdles of blood sample preparation. The devices will be categorized by application and the type of blood sample used. The final section focuses on devices for the detection of intracellular nucleic acids, because these require more extensive sample preparation steps, and the challenges involved in adapting this technology and potential improvements are discussed.
Collapse
Affiliation(s)
| | - Rena Hirani
- Australian Red Cross Lifeblood, Sydney, New South Wales 2015, Australia
| | - Andrew Care
- School of Life Sciences, University of Technology Sydney, Ultimo, New South Wales 2007, Australia
| | - David W Inglis
- School of Engineering, Faculty of Science and Engineering and △School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | | |
Collapse
|
31
|
Bhatti JS, Khullar N, Vijayvergiya R, Navik U, Bhatti GK, Reddy PH. Mitochondrial miRNA as epigenomic signatures: Visualizing aging-associated heart diseases through a new lens. Ageing Res Rev 2023; 86:101882. [PMID: 36780957 DOI: 10.1016/j.arr.2023.101882] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 02/05/2023] [Accepted: 02/08/2023] [Indexed: 02/13/2023]
Abstract
Aging bears many hard knocks, but heart disorders earn a particular allusion, being the most widespread. Cardiovascular diseases (CVDs) are becoming the biggest concern to mankind due to sundry health conditions directly or indirectly related to heart-linked abnormalities. Scientists know that mitochondria play a critical role in the pathophysiology of cardiac diseases. Both environment and genetics play an essential role in modulating and controlling mitochondrial functions. Even a minor abnormality may prove detrimental to heart function. Advanced age combined with an unhealthy lifestyle can cause most cardiomyocytes to be replaced by fibrotic tissue which upsets the conducting system and leads to arrhythmias. An aging heart encounters far more heart-associated comorbidities than a young heart. Many state-of-the-art technologies and procedures are already being used to prevent and treat heart attacks worldwide. However, it remains a mystery when this heart bomb would explode because it lacks an alarm. This calls for a novel and effective strategy for timely diagnosis and a sure-fire treatment. This review article provides a comprehensive overture of prospective potentials of mitochondrial miRNAs that predict complicated and interconnected pathways concerning heart ailments and signature compilations of relevant miRNAs as biomarkers to plot the role of miRNAs in epigenomics. This article suggests that analysis of DNA methylation patterns in age-associated heart diseases may determine age-impelled biomarkers of heart disease.
Collapse
Affiliation(s)
- Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India.
| | - Naina Khullar
- Department of Zoology, Mata Gujri College, Fatehgarh Sahib, Punjab, India.
| | - Rajesh Vijayvergiya
- Department of Cardiology, Post Graduate Institute of Medical Education and Research, Chandigarh, India.
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Bathinda, India.
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali, India.
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Neuroscience & Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Departments of Neurology, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA.
| |
Collapse
|
32
|
Bovari-Biri J, Garai K, Banfai K, Csongei V, Pongracz JE. miRNAs as Predictors of Barrier Integrity. BIOSENSORS 2023; 13:bios13040422. [PMID: 37185497 PMCID: PMC10136429 DOI: 10.3390/bios13040422] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 05/17/2023]
Abstract
The human body has several barriers that protect its integrity and shield it from mechanical, chemical, and microbial harm. The various barriers include the skin, intestinal and respiratory epithelia, blood-brain barrier (BBB), and immune system. In the present review, the focus is on the physical barriers that are formed by cell layers. The barrier function is influenced by the molecular microenvironment of the cells forming the barriers. The integrity of the barrier cell layers is maintained by the intricate balance of protein expression that is partly regulated by microRNAs (miRNAs) both in the intracellular space and the extracellular microenvironment. The detection of changes in miRNA patterns has become a major focus of diagnostic, prognostic, and disease progression, as well as therapy-response, markers using a great variety of detection systems in recent years. In the present review, we highlight the importance of liquid biopsies in assessing barrier integrity and challenges in differential miRNA detection.
Collapse
Affiliation(s)
- Judit Bovari-Biri
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, 2 Rokus Str, H-7624 Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, 20 Ifjusag Str, H-7624 Pecs, Hungary
| | - Kitti Garai
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, 2 Rokus Str, H-7624 Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, 20 Ifjusag Str, H-7624 Pecs, Hungary
| | - Krisztina Banfai
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, 2 Rokus Str, H-7624 Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, 20 Ifjusag Str, H-7624 Pecs, Hungary
| | - Veronika Csongei
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, 2 Rokus Str, H-7624 Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, 20 Ifjusag Str, H-7624 Pecs, Hungary
| | - Judit E Pongracz
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, 2 Rokus Str, H-7624 Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, 20 Ifjusag Str, H-7624 Pecs, Hungary
| |
Collapse
|
33
|
Berzina B, Peramune U, Kim S, Saurabh K, Claus EL, Strait ME, Ganapathysubramanian B, Anand RK. Electrokinetic Enrichment and Label-Free Electrochemical Detection of Nucleic Acids by Conduction of Ions along the Surface of Bioconjugated Beads. ACS Sens 2023; 8:1173-1182. [PMID: 36800317 DOI: 10.1021/acssensors.2c02480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
In this paper, we report a method to integrate the electrokinetic pre-enrichment of nucleic acids within a bed of probe-modified microbeads with their label-free electrochemical detection. In this detection scheme, hybridization of locally enriched target nucleic acids to the beads modulates the conduction of ions along the bead surfaces. This is a fundamental advancement in that this mechanism is similar to that observed in nanopore sensors, yet occurs in a bed of microbeads with microscale interstices. In application, this approach has several distinct advantages. First, electrokinetic enrichment requires only a simple DC power supply, and in combination with nonoptical detection, it makes this method amenable to point-of-care applications. Second, the sensor is easy to fabricate and comprises a packed bed of commercially available microbeads, which can be readily modified with a wide range of probe types, thereby making this a versatile platform. Finally, the sensor is highly sensitive (picomolar) despite the modest 100-fold pre-enrichment we employ here by faradaic ion concentration polarization (fICP). Further gains are anticipated under conditions for fICP focusing that are known to yield higher enrichment factors (up to 100,000-fold enrichment). Here, we demonstrate the detection of 3.7 pM single-stranded DNA complementary to the bead-bound oligoprobe, following a 30 min single step of enrichment and hybridization. Our results indicate that a shift in the slope of a current-voltage curve occurs upon hybridization and that this shift is proportional to the logarithm of the concentration of target DNA. Finally, we investigate the proposed mechanism of sensing by developing a numerical simulation that shows an increase in ion flux through the bed of insulating beads, given the changes in surface charge and zeta potential, consistent with our experimental conditions.
Collapse
Affiliation(s)
- Beatrise Berzina
- The Department of Chemistry, Iowa State University, 1605 Gilman Hall, 2415 Osborn Drive, Ames, Iowa 50011-1021, United States
| | - Umesha Peramune
- The Department of Chemistry, Iowa State University, 1605 Gilman Hall, 2415 Osborn Drive, Ames, Iowa 50011-1021, United States
| | - Sungu Kim
- The Department of Chemistry, Iowa State University, 1605 Gilman Hall, 2415 Osborn Drive, Ames, Iowa 50011-1021, United States
- The Department of Mechanical Engineering, Iowa State University, 2043 Black Engineering, 2529 Union Drive, Ames, Iowa 50011-2030, United States
| | - Kumar Saurabh
- The Department of Mechanical Engineering, Iowa State University, 2043 Black Engineering, 2529 Union Drive, Ames, Iowa 50011-2030, United States
| | - Echo L Claus
- The Department of Chemistry, Iowa State University, 1605 Gilman Hall, 2415 Osborn Drive, Ames, Iowa 50011-1021, United States
| | - Madison E Strait
- The Department of Chemistry, Iowa State University, 1605 Gilman Hall, 2415 Osborn Drive, Ames, Iowa 50011-1021, United States
| | - Baskar Ganapathysubramanian
- The Department of Mechanical Engineering, Iowa State University, 2043 Black Engineering, 2529 Union Drive, Ames, Iowa 50011-2030, United States
| | - Robbyn K Anand
- The Department of Chemistry, Iowa State University, 1605 Gilman Hall, 2415 Osborn Drive, Ames, Iowa 50011-1021, United States
| |
Collapse
|
34
|
Kumar S, Maniya N, Wang C, Senapati S, Chang HC. Quantifying PON1 on HDL with nanoparticle-gated electrokinetic membrane sensor for accurate cardiovascular risk assessment. Nat Commun 2023; 14:557. [PMID: 36732521 PMCID: PMC9895453 DOI: 10.1038/s41467-023-36258-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 01/23/2023] [Indexed: 02/04/2023] Open
Abstract
Cardiovascular disease-related deaths (one-third of global deaths) can be reduced with a simple screening test for better biomarkers than the current lipid and lipoprotein profiles. We propose using a highly atheroprotective subset of HDL with colocalized PON1 (PON1-HDL) for superior cardiovascular risk assessment. However, direct quantification of HDL proteomic subclasses are complicated by the peroxides/antioxidants associated with HDL interfering with redox reactions in enzymatic calorimetric and electrochemical immunoassays. Hence, we developed an enzyme-free Nanoparticle-Gated Electrokinetic Membrane Sensor (NGEMS) platform for quantification of PON1-HDL in plasma within 60 min, with a sub-picomolar limit of detection, 3-4 log dynamic range and without needing sample pretreatment or individual-sample calibration. Using NGEMS, we report our study on human plasma PON1-HDL as a cardiovascular risk marker with AUC~0.99 significantly outperforming others (AUC~0.6-0.8), including cholesterol/triglycerides tests. Validation for a larger cohort can establish PON1-HDL as a biomarker that can potentially reshape cardiovascular landscape.
Collapse
Affiliation(s)
- Sonu Kumar
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Indiana, USA
| | - Nalin Maniya
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Indiana, USA
| | - Ceming Wang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Indiana, USA
| | - Satyajyoti Senapati
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Indiana, USA.
| | - Hsueh-Chia Chang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Indiana, USA.
| |
Collapse
|
35
|
Li L, Zhang L, Montgomery KC, Jiang L, Lyon CJ, Hu TY. Advanced technologies for molecular diagnosis of cancer: State of pre-clinical tumor-derived exosome liquid biopsies. Mater Today Bio 2023; 18:100538. [PMID: 36619206 PMCID: PMC9812720 DOI: 10.1016/j.mtbio.2022.100538] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
Exosomes are membrane-defined extracellular vesicles (EVs) approximately 40-160 nm in diameter that are found in all body fluids including blood, urine, and saliva. They act as important vehicles for intercellular communication between both local and distant cells and can serve as circulating biomarkers for disease diagnosis and prognosis. Exosomes play a key role in tumor metastasis, are abundant in biofluids, and stabilize biomarkers they carry, and thus can improve cancer detection, treatment monitoring, and cancer staging/prognosis. Despite their clinical potential, lack of sensitive/specific biomarkers and sensitive isolation/enrichment and analytical technologies has posed a barrier to clinical translation of exosomes. This review presents a critical overview of technologies now being used to detect tumor-derived exosome (TDE) biomarkers in clinical specimens that have potential for clinical translation.
Collapse
Affiliation(s)
- Lin Li
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
- Center for Cellular and Molecular Diagnostics, Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Lili Zhang
- Center for Cellular and Molecular Diagnostics, Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, LA, USA
- HCA Florida Healthcare Westside/Northwest Hospital Internal Medicine, Plantation, Florida, USA
| | - Katelynn C. Montgomery
- Department of Biomedical Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, USA
| | - Li Jiang
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Christopher J. Lyon
- Center for Cellular and Molecular Diagnostics, Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Tony Y. Hu
- Center for Cellular and Molecular Diagnostics, Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, LA, USA
- Department of Biomedical Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, USA
| |
Collapse
|
36
|
Ma Y, Liu C, Cao S, Chen T, Chen G. Microfluidics for diagnosis and treatment of cardiovascular disease. J Mater Chem B 2023; 11:546-559. [PMID: 36542463 DOI: 10.1039/d2tb02287g] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease (CVD), a type of circulatory system disease related to the lesions of the cardiovascular system, has become one of the main diseases that endanger human health. Currently, the clinical diagnosis of most CVDs relies on a combination of imaging technology and blood biochemical test. However, the existing technologies for diagnosis of CVDs still have limitations in terms of specificity, detection range, and cost. In order to break through the current bottleneck, microfluidic with the advantages of low cost, simple instruments and easy integration, has been developed to play an important role in the early prevention, diagnosis and treatment of CVDs. Here, we have reviewed the recent various applications of microfluidic in the clinical diagnosis and treatment of CVDs, including microfluidic devices for detecting CVD markers, the cardiovascular models based on microfluidic, and the microfluidic used for CVDs drug screening and delivery. In addition, we have briefly looked forward to the prospects and challenges of microfluidics in diagnosis and treatment of CVDs.
Collapse
Affiliation(s)
- Yonggeng Ma
- School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Chenbin Liu
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, P. R. China
| | - Siyu Cao
- School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Tianshu Chen
- Department of Clinical Laboratory Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China.
| | - Guifang Chen
- School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| |
Collapse
|
37
|
McCarthy KP, Go DB, Senapati S, Chang HC. An integrated ion-exchange membrane-based microfluidic device for irreversible dissociation and quantification of miRNA from ribonucleoproteins. LAB ON A CHIP 2023; 23:285-294. [PMID: 36524732 PMCID: PMC10697430 DOI: 10.1039/d2lc00517d] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Ribonucleoproteins (RNPs), particularly microRNA-induced silencing complex (miRISC), have been associated with cancer-related gene regulation. Specific RNA-protein associations in miRISC complexes or those found in let-7 lin28A complexes can downregulate tumor-suppressing genes and can be directly linked to cancer. The high protein-RNA electrostatic binding affinity is a particular challenge for the quantification of the associated microRNAs (miRNAs). We report here the first microfluidic point-of-care assay that allows direct quantification of RNP-associated RNAs, which has the potential to greatly advance RNP profiling for liquid biopsy. Key to the technology is an integrated cation-anion exchange membrane (CEM/AEM) platform for rapid and irreversible dissociation (k = 0.0025 s-1) of the RNP (Cas9-miR-21) complex and quantification of its associated miR-21 in 40 minutes. The CEM-induced depletion front is used to concentrate the RNP at the depletion front such that the high electric field (>100 V cm-1) within the concentration boundary layer induces irreversible dissociation of the low KD (∼0.5 nM) complex, with ∼100% dissociation even though the association rate (kon = 6.1 s-1) is 1000 times higher. The high field also electrophoretically drives the dissociated RNA out of the concentrated zone without reassociation. A detection limit of 1.1 nM is achieved for Cy3 labelled miR-21.
Collapse
Affiliation(s)
- Kyle P McCarthy
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA.
| | - David B Go
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA.
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Satyajyoti Senapati
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA.
| | - Hsueh-Chia Chang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA.
| |
Collapse
|
38
|
Wang Y, Wang S, Li L, Zou Y, Liu B, Fang X. Microfluidics‐based molecular profiling of tumor‐derived exosomes for liquid biopsy. VIEW 2023. [DOI: 10.1002/viw.20220048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Affiliation(s)
- Yuqing Wang
- School of Pharmacy Shanghai Stomatological Hospital Department of Chemistry Fudan University Shanghai China
| | - Shurong Wang
- School of Pharmacy Shanghai Stomatological Hospital Department of Chemistry Fudan University Shanghai China
| | - Lanting Li
- School of Pharmacy Shanghai Stomatological Hospital Department of Chemistry Fudan University Shanghai China
| | - Yan Zou
- School of Pharmacy Shanghai Stomatological Hospital Department of Chemistry Fudan University Shanghai China
| | - Baohong Liu
- School of Pharmacy Shanghai Stomatological Hospital Department of Chemistry Fudan University Shanghai China
| | - Xiaoni Fang
- School of Pharmacy Shanghai Stomatological Hospital Department of Chemistry Fudan University Shanghai China
| |
Collapse
|
39
|
Azeem MM, Shafa M, Aamir M, Zubair M, Souayeh B, Alam MW. Nucleotide detection mechanism and comparison based on low-dimensional materials: A review. Front Bioeng Biotechnol 2023; 11:1117871. [PMID: 36937765 PMCID: PMC10018150 DOI: 10.3389/fbioe.2023.1117871] [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: 12/07/2022] [Accepted: 02/13/2023] [Indexed: 03/06/2023] Open
Abstract
The recent pandemic has led to the fabrication of new nucleic acid sensors that can detect infinitesimal limits immediately and effectively. Therefore, various techniques have been demonstrated using low-dimensional materials that exhibit ultrahigh detection and accuracy. Numerous detection approaches have been reported, and new methods for impulse sensing are being explored. All ongoing research converges at one unique point, that is, an impetus: the enhanced limit of detection of sensors. There are several reviews on the detection of viruses and other proteins related to disease control point of care; however, to the best of our knowledge, none summarizes the various nucleotide sensors and describes their limits of detection and mechanisms. To understand the far-reaching impact of this discipline, we briefly discussed conventional and nanomaterial-based sensors, and then proposed the feature prospects of these devices. Two types of sensing mechanisms were further divided into their sub-branches: polymerase chain reaction and photospectrometric-based sensors. The nanomaterial-based sensor was further subdivided into optical and electrical sensors. The optical sensors included fluorescence (FL), surface plasmon resonance (SPR), colorimetric, and surface-enhanced Raman scattering (SERS), while electrical sensors included electrochemical luminescence (ECL), microfluidic chip, and field-effect transistor (FET). A synopsis of sensing materials, mechanisms, detection limits, and ranges has been provided. The sensing mechanism and materials used were discussed for each category in terms of length, collectively forming a fusing platform to highlight the ultrahigh detection technique of nucleotide sensors. We discussed potential trends in improving the fabrication of nucleotide nanosensors based on low-dimensional materials. In this area, particular aspects, including sensitivity, detection mechanism, stability, and challenges, were addressed. The optimization of the sensing performance and selection of the best sensor were concluded. Recent trends in the atomic-scale simulation of the development of Deoxyribonucleic acid (DNA) sensors using 2D materials were highlighted. A critical overview of the challenges and opportunities of deoxyribonucleic acid sensors was explored, and progress made in deoxyribonucleic acid detection over the past decade with a family of deoxyribonucleic acid sensors was described. Areas in which further research is needed were included in the future scope.
Collapse
Affiliation(s)
- M. Mustafa Azeem
- Department of Civil, Architectural, and Environmental Engineering, Missouri University of Science and Technology, Rolla, MO, United States
- *Correspondence: M. Mustafa Azeem, ; Muhammad Aamir,
| | - Muhammad Shafa
- Yunnan Key Laboratory of Metal-Organic Molecular Materials and Devices, Kunming University, Kunming, Yunnan, China
| | - Muhammad Aamir
- Department of Basic Science, Deanship of Preparatory Year, King Faisal University, Hofuf, Saudi Arabia
- *Correspondence: M. Mustafa Azeem, ; Muhammad Aamir,
| | - Muhammad Zubair
- Mechanical and Nuclear Engineering Department, University of Sharjah, Sharjah, United Arab Emirates
| | - Basma Souayeh
- Department of Physics, College of Science, King Faisal University, Al Ahsa, Saudi Arabia
| | - Mir Waqas Alam
- Department of Physics, College of Science, King Faisal University, Al Ahsa, Saudi Arabia
| |
Collapse
|
40
|
Abstract
Exosomes are extracellular vesicles, which have the ability to convey various types of cargo between cells. Lately, a great amount of interest has been paid to exosomal microRNAs (miRNAs), since much evidence has suggested that the sorting of miRNAs into exosomes is not an accidental process. It has been shown that exosomal miRNAs (exo-miRNAs) are implicated in a variety of cellular processes including (but not limited to) cell migration, apoptosis, proliferation, and autophagy. Exosomes can play a role in cardiovascular diseases and can be used as diagnostic biomarkers for several diseases, especially cancer. Tremendous advances in technology have led to the development of various platforms for miRNA profiling. Each platform has its own limitations and strengths that need to be understood in order to use them properly. In the current review, we summarize some exo-miRNAs that are relevant to exo-miRNA profiling studies and describe new methods used for the measurement of miRNA profiles in different human bodily fluids.
Collapse
|
41
|
Kumar K, Kim E, Alhammadi M, Umapathi R, Aliya S, Tiwari JN, Park HS, Choi JH, Son CY, Vilian AE, Han YK, Bu J, Huh YS. Recent advances in microfluidic approaches for the isolation and detection of exosomes. Trends Analyt Chem 2023. [DOI: 10.1016/j.trac.2022.116912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
42
|
Visan KS, Wu LY, Voss S, Wuethrich A, Möller A. Status quo of Extracellular Vesicle isolation and detection methods for clinical utility. Semin Cancer Biol 2023; 88:157-171. [PMID: 36581020 DOI: 10.1016/j.semcancer.2022.12.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/20/2022] [Accepted: 12/25/2022] [Indexed: 12/28/2022]
Abstract
Extracellular vesicles (EVs) are nano-sized particles that hold tremendous potential in the clinical space, as their biomolecular profiles hold a key to non-invasive liquid biopsy for cancer diagnosis and prognosis. EVs are present in most bodily fluids, hence are easily obtainable from patients, advantageous to that of traditional, invasive tissue biopsies and imaging techniques. However, there are certain constraints that hinder clinical use of EVs. The translation of EV biomarkers from "bench-to-bedside" is encumbered by the methods of EV isolation and subsequent biomarker detection currently implemented in laboratories. Although current isolation and detection methods are effective, they lack practicality, with their requirement for high bodily fluid volumes, low equipment availability, slow turnaround times and high costs. The high demand for techniques that overcome these limitations has resulted in significant advancements in nanotechnological devices. These devices are designed to integrate EV isolation and biomarker detection into a one-step method of direct EV detection from bodily fluids. This provides promise for the acceleration of EVs into current clinical standards. This review highlights the importance of EVs as cancer biomarkers, the methodological obstacles currently faced in clinical studies and how novel nanodevices could advance clinical translation.
Collapse
Affiliation(s)
- Kekoolani S Visan
- Tumour Microenvironment Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; Department of Otorhinolaryngology, Head and Neck Surgery, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Li-Ying Wu
- Tumour Microenvironment Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; Department of Otorhinolaryngology, Head and Neck Surgery, Chinese University of Hong Kong, Shatin, Hong Kong; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland 4059, Australia
| | - Sarah Voss
- Tumour Microenvironment Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; Department of Otorhinolaryngology, Head and Neck Surgery, Chinese University of Hong Kong, Shatin, Hong Kong; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland 4059, Australia
| | - Alain Wuethrich
- Centre for Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Andreas Möller
- Tumour Microenvironment Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; Department of Otorhinolaryngology, Head and Neck Surgery, Chinese University of Hong Kong, Shatin, Hong Kong.
| |
Collapse
|
43
|
Khashayar P, Al-Madhagi S, Azimzadeh M, Scognamiglio V, Arduini F. New frontiers in microfluidics devices for miRNA analysis. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116706] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
44
|
Xu H, Ye BC. Integrated microfluidic platforms for tumor-derived exosome analysis. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
45
|
Kolmar L, Autour A, Ma X, Vergier B, Eduati F, Merten CA. Technological and computational advances driving high-throughput oncology. Trends Cell Biol 2022; 32:947-961. [PMID: 35577671 DOI: 10.1016/j.tcb.2022.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/11/2022] [Accepted: 04/20/2022] [Indexed: 01/21/2023]
Abstract
Engineering and computational advances have opened many new avenues in cancer research, particularly when being exploited in interdisciplinary approaches. For example, the combination of microfluidics, novel sequencing technologies, and computational analyses has been crucial to enable single-cell assays, giving a detailed picture of tumor heterogeneity for the very first time. In a similar way, these 'tech' disciplines have been elementary for generating large data sets in multidimensional cancer 'omics' approaches, cell-cell interaction screens, 3D tumor models, and tissue level analyses. In this review we summarize the most important technology and computational developments that have been or will be instrumental for transitioning classical cancer research to a large data-driven, high-throughput, high-content discipline across all biological scales.
Collapse
Affiliation(s)
- Leonie Kolmar
- Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Alexis Autour
- Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland; European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Xiaoli Ma
- Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Blandine Vergier
- Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Federica Eduati
- Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands.
| | - Christoph A Merten
- Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
46
|
Glass SE, Coffey RJ. Recent Advances in the Study of Extracellular Vesicles in Colorectal Cancer. Gastroenterology 2022; 163:1188-1197. [PMID: 35724732 PMCID: PMC9613516 DOI: 10.1053/j.gastro.2022.06.039] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/08/2022] [Accepted: 06/11/2022] [Indexed: 12/15/2022]
Abstract
There has been significant progress in the study of extracellular vesicles (EVs) since the 2017 American Gastroenterological Association-sponsored Freston Conference "Extracellular Vesicles: Biology, Translation and Clinical Application in GI Disorders." The burgeoning interest in this field stems from the increasing recognition that EVs represent an understudied form of cell-to-cell communication and contain cargo replete with biomarkers and therapeutic targets. This short review will highlight recent advances in the field, with an emphasis on colorectal cancer. After a brief introduction to secreted particles, we will describe how our laboratory became interested in EVs, which led to refined methods of isolation and identification of 2 secreted nanoparticles. We will then summarize the cargo found in small EVs released from colorectal cancer cells and other cells in the tumor microenvironment, as well as those found in the circulation of patients with colorectal cancer. Finally, we will consider the continuing challenges and future opportunities in this rapidly evolving field.
Collapse
Affiliation(s)
- Sarah E Glass
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Robert J Coffey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
47
|
Yin S, Chen A, Ding Y, Song J, Chen R, Zhang P, Yang C. Recent advances in exosomal RNAs analysis towards diagnostic and therapeutic applications. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
48
|
Lin B, Jiang J, Jia J, Zhou X. Recent Advances in Exosomal miRNA Biosensing for Liquid Biopsy. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27217145. [PMID: 36363975 PMCID: PMC9655350 DOI: 10.3390/molecules27217145] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 12/05/2022]
Abstract
As a noninvasive detection technique, liquid biopsy plays a valuable role in cancer diagnosis, disease monitoring, and prognostic assessment. In liquid biopsies, exosomes are considered among the potential biomarkers because they are important bioinformation carriers for intercellular communication. Exosomes transport miRNAs and, thus, play an important role in the regulation of cell growth and function; therefore, detection of cancer cell-derived exosomal miRNAs (exo-miRNAs) gives effective information in liquid biopsy. The development of sensitive, convenient, and reliable exo-miRNA assays will provide new perspectives for medical diagnosis. This review presents different designs and detection strategies of recent exo-miRNA assays in terms of signal transduction and amplification, as well as signal detection. In addition, this review outlines the current attempts at bioassay methods in liquid biopsies. Lastly, the challenges and prospects of exosome bioassays are also considered.
Collapse
Affiliation(s)
- Bingqian Lin
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
- Correspondence: (B.L.); (X.Z.)
| | - Jinting Jiang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Jingxuan Jia
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Xiang Zhou
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
- Correspondence: (B.L.); (X.Z.)
| |
Collapse
|
49
|
Morales RTT, Ko J. Future of Digital Assays to Resolve Clinical Heterogeneity of Single Extracellular Vesicles. ACS NANO 2022; 16:11619-11645. [PMID: 35904433 PMCID: PMC10174080 DOI: 10.1021/acsnano.2c04337] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Extracellular vesicles (EVs) are complex lipid membrane vehicles with variable expressions of molecular cargo, composed of diverse subpopulations that participate in the intercellular signaling of biological responses in disease. EV-based liquid biopsies demonstrate invaluable clinical potential for overhauling current practices of disease management. Yet, EV heterogeneity is a major needle-in-a-haystack challenge to translate their use into clinical practice. In this review, existing digital assays will be discussed to analyze EVs at a single vesicle resolution, and future opportunities to optimize the throughput, multiplexing, and sensitivity of current digital EV assays will be highlighted. Furthermore, this review will outline the challenges and opportunities that impact the clinical translation of single EV technologies for disease diagnostics and treatment monitoring.
Collapse
Affiliation(s)
- Renee-Tyler T Morales
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Jina Ko
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
50
|
Mateescu B, Jones JC, Alexander RP, Alsop E, An JY, Asghari M, Boomgarden A, Bouchareychas L, Cayota A, Chang HC, Charest A, Chiu DT, Coffey RJ, Das S, De Hoff P, deMello A, D’Souza-Schorey C, Elashoff D, Eliato KR, Franklin JL, Galas DJ, Gerstein MB, Ghiran IH, Go DB, Gould S, Grogan TR, Higginbotham JN, Hladik F, Huang TJ, Huo X, Hutchins E, Jeppesen DK, Jovanovic-Talisman T, Kim BY, Kim S, Kim KM, Kim Y, Kitchen RR, Knouse V, LaPlante EL, Lebrilla CB, Lee LJ, Lennon KM, Li G, Li F, Li T, Liu T, Liu Z, Maddox AL, McCarthy K, Meechoovet B, Maniya N, Meng Y, Milosavljevic A, Min BH, Morey A, Ng M, Nolan J, De Oliveira Junior GP, Paulaitis ME, Phu TA, Raffai RL, Reátegui E, Roth ME, Routenberg DA, Rozowsky J, Rufo J, Senapati S, Shachar S, Sharma H, Sood AK, Stavrakis S, Stürchler A, Tewari M, Tosar JP, Tucker-Schwartz AK, Turchinovich A, Valkov N, Van Keuren-Jensen K, Vickers KC, Vojtech L, Vreeland WN, Wang C, Wang K, Wang Z, Welsh JA, Witwer KW, Wong DT, Xia J, Xie YH, Yang K, Zaborowski MP, Zhang C, Zhang Q, Zivkovic AM, Laurent LC. Phase 2 of extracellular RNA communication consortium charts next-generation approaches for extracellular RNA research. iScience 2022; 25:104653. [PMID: 35958027 PMCID: PMC9358052 DOI: 10.1016/j.isci.2022.104653] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The extracellular RNA communication consortium (ERCC) is an NIH-funded program aiming to promote the development of new technologies, resources, and knowledge about exRNAs and their carriers. After Phase 1 (2013-2018), Phase 2 of the program (ERCC2, 2019-2023) aims to fill critical gaps in knowledge and technology to enable rigorous and reproducible methods for separation and characterization of both bulk populations of exRNA carriers and single EVs. ERCC2 investigators are also developing new bioinformatic pipelines to promote data integration through the exRNA atlas database. ERCC2 has established several Working Groups (Resource Sharing, Reagent Development, Data Analysis and Coordination, Technology Development, nomenclature, and Scientific Outreach) to promote collaboration between ERCC2 members and the broader scientific community. We expect that ERCC2's current and future achievements will significantly improve our understanding of exRNA biology and the development of accurate and efficient exRNA-based diagnostic, prognostic, and theranostic biomarker assays.
Collapse
Affiliation(s)
- Bogdan Mateescu
- Brain Research Institute, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
- Institute for Chemical and Bioengineering, ETH Zürich, Vladimir Prelog Weg 1, 8093 Zürich, Switzerland
| | - Jennifer C. Jones
- Laboratory of Pathology Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Eric Alsop
- Neurogenomics Division, TGen, Phoenix, AZ 85004, USA
| | - Ji Yeong An
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Mohammad Asghari
- Institute for Chemical and Bioengineering, ETH Zürich, Vladimir Prelog Weg 1, 8093 Zürich, Switzerland
| | - Alex Boomgarden
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Laura Bouchareychas
- Department of Surgery, Division of Vascular and Endovascular Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- Northern California Institute for Research and Education, San Francisco, CA 94121, USA
| | - Alfonso Cayota
- Functional Genomics Unit, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
- University Hospital, Universidad de la República, Montevideo 11600, Uruguay
| | - Hsueh-Chia Chang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Al Charest
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Daniel T. Chiu
- Department of Chemistry and Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Robert J. Coffey
- Department of Medicine/Gastroenterology and Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37235, USA
| | - Saumya Das
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Peter De Hoff
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, La Jolla, San Diego, CA 92093, USA
| | - Andrew deMello
- Institute for Chemical and Bioengineering, ETH Zürich, Vladimir Prelog Weg 1, 8093 Zürich, Switzerland
| | | | - David Elashoff
- Statistics Core, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Kiarash R. Eliato
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Jeffrey L. Franklin
- Department of Medicine/Gastroenterology and Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37235, USA
| | - David J. Galas
- Pacific Northwest Research Institute, Seattle, WA 98122, USA
| | - Mark B. Gerstein
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, CT 06520, USA
- Department of Computer Science, Yale University, New Haven, CT 06520, USA
| | - Ionita H. Ghiran
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - David B. Go
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Stephen Gould
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Tristan R. Grogan
- Department of Medicine Statistics Core, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - James N. Higginbotham
- Department of Medicine/Gastroenterology and Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Florian Hladik
- Departments of Obstetrics and Gynecology, and Medicine, University of Washington, Seattle, WA, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Tony Jun Huang
- Department of Mechanical Engineering and Material Science, Duke University, Durham, NC 27708, USA
| | - Xiaoye Huo
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | | | - Dennis K. Jeppesen
- Department of Medicine/Gastroenterology and Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Tijana Jovanovic-Talisman
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Betty Y.S. Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sung Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kyoung-Mee Kim
- Department of Pathology & Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yong Kim
- Department of Oral Biology and Medicine, UCLA School of Dentistry, Los Angeles, CA 90095, USA
| | - Robert R. Kitchen
- Corrigan Minehan Heart Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Vaughan Knouse
- Laboratory of Pathology Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Emily L. LaPlante
- Bioinformatics Research Laboratory, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - L. James Lee
- Department of Chemical and Biomolecular Engineering and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Kathleen M. Lennon
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Guoping Li
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Feng Li
- Department of Oral Biology and Medicine, UCLA School of Dentistry, Los Angeles, CA 90095, USA
| | - Tieyi Li
- Department of Materials Science & Engineering, University of California Los Angeles, Los Angeles, CA 90095-1595, USA
| | - Tao Liu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Zirui Liu
- Department of Materials Science & Engineering, University of California Los Angeles, Los Angeles, CA 90095-1595, USA
| | - Adam L. Maddox
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Kyle McCarthy
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | | | - Nalin Maniya
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Yingchao Meng
- Institute for Chemical and Bioengineering, ETH Zürich, Vladimir Prelog Weg 1, 8093 Zürich, Switzerland
| | - Aleksandar Milosavljevic
- Bioinformatics Research Laboratory, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Program in Quantitative and Computational Biosciences Baylor College of Medicine, Houston, TX 77030, USA
| | - Byoung-Hoon Min
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University, School of Medicine, Seoul, South Korea
| | - Amber Morey
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, La Jolla, San Diego, CA 92093, USA
| | - Martin Ng
- Northern California Institute for Research and Education, San Francisco, CA 94121, USA
| | - John Nolan
- Scintillon Institute, San Diego, CA, USA
| | | | - Michael E. Paulaitis
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Tuan Anh Phu
- Northern California Institute for Research and Education, San Francisco, CA 94121, USA
| | - Robert L. Raffai
- Department of Surgery, Division of Vascular and Endovascular Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- Northern California Institute for Research and Education, San Francisco, CA 94121, USA
- Department of Veterans Affairs, Surgical Service (112G), San Francisco VA Medical Center, San Francisco, CA 94121, USA
| | - Eduardo Reátegui
- Department of Chemical and Biomolecular Engineering and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Matthew E. Roth
- Bioinformatics Research Laboratory, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Joel Rozowsky
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA
| | - Joseph Rufo
- Department of Mechanical Engineering and Material Science, Duke University, Durham, NC 27708, USA
| | - Satyajyoti Senapati
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Sigal Shachar
- Meso Scale Diagnostics, LLC, Rockville, MD 20850, USA
| | - Himani Sharma
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Anil K. Sood
- Department of Gynecologic Oncology & Reproductive Medicine, University of Texas MD Aderson Cancer Center, Houston, TX 77030, USA
| | - Stavros Stavrakis
- Institute for Chemical and Bioengineering, ETH Zürich, Vladimir Prelog Weg 1, 8093 Zürich, Switzerland
| | - Alessandra Stürchler
- Brain Research Institute, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
- Institute for Chemical and Bioengineering, ETH Zürich, Vladimir Prelog Weg 1, 8093 Zürich, Switzerland
| | - Muneesh Tewari
- Department of Internal Medicine, Hematology/Oncology Division, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Juan P. Tosar
- Functional Genomics Unit, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
- Analytical Biochemistry Unit, School of Science, Universidad de la República, Montevideo 11400, Uruguay
| | | | - Andrey Turchinovich
- Cancer Genome Research (B063), German Cancer Research Center DKFZ, Heidelberg 69120, Germany
- Heidelberg Biolabs GmbH, Heidelberg 69120, Germany
| | - Nedyalka Valkov
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | | | - Kasey C. Vickers
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lucia Vojtech
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Wyatt N. Vreeland
- Bioprocess Measurement Group, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - Ceming Wang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Kai Wang
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - ZeYu Wang
- Department of Mechanical Engineering and Material Science, Duke University, Durham, NC 27708, USA
| | - Joshua A. Welsh
- Laboratory of Pathology Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Kenneth W. Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - David T.W. Wong
- Department of Oral Biology and Medicine, UCLA School of Dentistry, Los Angeles, CA 90095, USA
| | - Jianping Xia
- Department of Mechanical Engineering and Material Science, Duke University, Durham, NC 27708, USA
| | - Ya-Hong Xie
- Department of Materials Science & Engineering, University of California Los Angeles, Los Angeles, CA 90095-1595, USA
| | - Kaichun Yang
- Department of Mechanical Engineering and Material Science, Duke University, Durham, NC 27708, USA
| | - Mikołaj P. Zaborowski
- Department of Gynecology, Obstetrics and Gynecologic Oncology, Division of Gynecologic Oncology, Poznan University of Medical Sciences, 60-535 Poznań, Poland
| | - Chenguang Zhang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Qin Zhang
- Department of Medicine/Gastroenterology and Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | - Louise C. Laurent
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, La Jolla, San Diego, CA 92093, USA
| |
Collapse
|