1
|
Di Santo R, Verdelli F, Niccolini B, Varca S, Gaudio AD, Di Giacinto F, De Spirito M, Pea M, Giovine E, Notargiacomo A, Ortolani M, Di Gaspare A, Baldi A, Pizzolante F, Ciasca G. Exploring novel circulating biomarkers for liver cancer through extracellular vesicle characterization with infrared spectroscopy and plasmonics. Anal Chim Acta 2024; 1319:342959. [PMID: 39122286 DOI: 10.1016/j.aca.2024.342959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 06/16/2024] [Accepted: 07/07/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the most common form of liver cancer, with cirrhosis being a major risk factor. Traditional blood markers like alpha-fetoprotein (AFP) demonstrate limited efficacy in distinguishing between HCC and cirrhosis, underscoring the need for more effective diagnostic methodologies. In this context, extracellular vesicles (EVs) have emerged as promising candidates; however, their practical diagnostic application is restricted by the current lack of label-free methods to accurately profile their molecular content. To address this gap, our study explores the potential of mid-infrared (mid-IR) spectroscopy, both alone and in combination with plasmonic nanostructures, to detect and characterize circulating EVs. RESULTS EVs were extracted from HCC and cirrhotic patients. Mid-IR spectroscopy in the Attenuated Total Reflection (ATR) mode was utilized to identify potential signatures for patient classification, highlighting significant changes in the Amide I-II region (1475-1700 cm-1). This signature demonstrated diagnostic performance comparable to AFP and surpassed it when the two markers were combined. Further investigations utilized a plasmonic metasurface suitable for ultrasensitive spectroscopy within this spectral range. This device consists of two sets of parallel rod-shaped gold nanoantennas (NAs); the longer NAs produced an intense near-field amplification in the Amide I-II bands, while the shorter NAs were utilized to provide a sharp reflectivity edge at 1800-2200 cm-1 for EV mass-sensing. A clinically relevant subpopulation of EVs was targeted by conjugating NAs with an antibody specific to Epithelial Cell Adhesion Molecule (EpCAM). This methodology enabled the detection of variations in the quantity of EpCAM-presenting EVs and revealed changes in the Amide I-II lineshape. SIGNIFICANCE The presented results can positively impact the development of novel laboratory methods for the label-free characterization of EVs, based on the combination between mid-IR spectroscopy and plasmonics. Additionally, data obtained by using HCC and cirrhotic subjects as a model system, suggest that this approach could be adapted for monitoring these conditions.
Collapse
Affiliation(s)
- R Di Santo
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore & Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Rome, Italy; Dipartimento di Scienze della Vita, della salute e delle Professioni sanitarie, Link Campus University, Rome, Italy
| | - F Verdelli
- Dutch Institute for Fundamental Energy Research (DIFFER), Eindhoven 5600 HH, The Netherlands
| | - B Niccolini
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore & Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Rome, Italy
| | - S Varca
- UOC of Gastroenterology, Department of Medical and Surgery Sciences, Fondazione Policlinico Universitario A.Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - A Del Gaudio
- UOC of Gastroenterology, Department of Medical and Surgery Sciences, Fondazione Policlinico Universitario A.Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - F Di Giacinto
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore & Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Rome, Italy
| | - M De Spirito
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore & Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Rome, Italy.
| | - M Pea
- Istituto di Fotonica e Nanotecnologie, Consiglio Nazionale delle Ricerche IFN-CNR, Via Del Fosso Del Cavaliere 100, 00133, Rome, Italy
| | - E Giovine
- Istituto di Fotonica e Nanotecnologie, Consiglio Nazionale delle Ricerche IFN-CNR, Via Del Fosso Del Cavaliere 100, 00133, Rome, Italy
| | - A Notargiacomo
- Istituto di Fotonica e Nanotecnologie, Consiglio Nazionale delle Ricerche IFN-CNR, Via Del Fosso Del Cavaliere 100, 00133, Rome, Italy
| | - M Ortolani
- Department of Physics, Sapienza University of Rome, Piazzale Aldo Moro 2, 00185, Rome, Italy
| | - A Di Gaspare
- NEST, CNR - Istituto Nanoscienze and Scuola Normale Superiore, Piazza San Silvestro 12, 56127, Pisa, Italy
| | - A Baldi
- Department of Physics and Astronomy, Vrije Universiteit Amsterdam, De Boelelaan 1081, 1081 HV Amsterdam, Netherlands
| | - F Pizzolante
- UOC of Gastroenterology, Department of Medical and Surgery Sciences, Fondazione Policlinico Universitario A.Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - G Ciasca
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore & Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Rome, Italy.
| |
Collapse
|
2
|
Romanò S, Nele V, Campani V, De Rosa G, Cinti S. A comprehensive guide to extract information from extracellular vesicles: a tutorial review towards novel analytical developments. Anal Chim Acta 2024; 1302:342473. [PMID: 38580402 DOI: 10.1016/j.aca.2024.342473] [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: 01/03/2024] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 04/07/2024]
Abstract
In the medical field, extracellular vesicles (EVs) are gaining importance as they act as cells mediators. These are phospholipid bilayer vesicles and contain crucial biochemical information about their mother cells being carrier of different biomolecules such as small molecules, proteins, lipids, and nucleic acids. After release into the extracellular matrix, they enter the systemic circulation and can be found in all human biofluids. Since EVs reflect the state of the cell of origin, there is exponential attention as potential source of new circulating biomarkers for liquid biopsy. The use of EVs in clinical practice faces several challenges that need to be addressed: these include the standardization of lysis protocols, the availability of low-cost reagents and the development of analytical tools capable of detecting biomarkers. The process of lysis is a crucial step that can impact all subsequent analyses, towards the development of novel analytical strategies. To aid researchers to support the evolution of measurement science technology, this tutorial review evaluates and discuss the most commonly protocols used to characterize the contents of EVs, including their advantages and disadvantages in terms of experimental procedures, time and equipment. The purpose of this tutorial review is to offer practical guide to researchers which are intended to develop novel analytical approaches. Some of the most significant applications are considered, highlighting their main characteristics divided per mechanism of action. Finally, comprehensive tables which provide an overview at a glance are provided to readers.
Collapse
Affiliation(s)
- Sabrina Romanò
- Department of Pharmacy, University of Naples Federico II, Italy.
| | - Valeria Nele
- Department of Pharmacy, University of Naples Federico II, Italy
| | | | | | - Stefano Cinti
- Department of Pharmacy, University of Naples Federico II, Italy.
| |
Collapse
|
3
|
Mishra A, Zehra S, Bharti PK, Mathur SR, Ranjan P, Batra A, Inampudi KK, Modi GP, Nikolajeff F, Kumar S. Spectroscopic insight into breast cancer: profiling small extracellular vesicles lipids via infrared spectroscopy for diagnostic precision. Sci Rep 2024; 14:9347. [PMID: 38654096 DOI: 10.1038/s41598-024-59863-1] [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: 12/05/2023] [Accepted: 04/16/2024] [Indexed: 04/25/2024] Open
Abstract
Breast cancer, a leading cause of female mortality due to delayed detection owing to asymptomatic nature and limited early diagnostic tools, was investigated using a multi-modal approach. Plasma-derived small EVs from breast cancer patients (BrCa, n = 74) and healthy controls (HC, n = 30) were analyzed. Small EVs (n = 104), isolated through chemical precipitation, underwent characterization via transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA). Validation involved antibody-based tests (TSG101, CD9, CD81, CD63). Infrared spectra of small EVs were obtained, revealing significant differences in lipid acyl chains, particularly in the C-H stretching of CH3. The study focused on the lipid region (3050-2900 cm-1), identifying peaks (3015 cm-1, 2960 cm-1, 2929 cm-1) as distinctive lipid characteristics. Spectroscopic lipid-to-lipid ratios [(I3015/I2929), (I2960/I2929)] emerged as prominent breast cancer markers. Exploration of protein, nucleic acid, and carbohydrate ratios indicated variations in alpha helices, asymmetric C-H stretching vibrations, and C-O stretching at 1033 cm-1. Principal component analysis (PCA) successfully differentiated BrCa and HC small EVs, and heatmap analysis and receiver operating characteristic (ROC) curve evaluations underscored the discriminatory power of lipid ratios. Notably, (I2960/I2929) exhibited 100% sensitivity and specificity, highlighting its potential as a robust BrCa sEV marker for breast cancer detection.
Collapse
Affiliation(s)
- Abhay Mishra
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Sadaqa Zehra
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Prahalad Kumar Bharti
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Sandeep R Mathur
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Piyush Ranjan
- Department of Surgical Disciplines, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Atul Batra
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Krishna K Inampudi
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Gyan Prakash Modi
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, Uttar Pradesh, India
| | - Fredrik Nikolajeff
- Department of Health, Education, and Technology, Lulea University of Technology, 97187, Luleå, Sweden
| | - Saroj Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India.
- Department of Health, Education, and Technology, Lulea University of Technology, 97187, Luleå, Sweden.
| |
Collapse
|
4
|
Medina-Ramirez IE, Macias-Diaz JE, Masuoka-Ito D, Zapien JA. Holotomography and atomic force microscopy: a powerful combination to enhance cancer, microbiology and nanotoxicology research. DISCOVER NANO 2024; 19:64. [PMID: 38594446 PMCID: PMC11003950 DOI: 10.1186/s11671-024-04003-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/23/2024] [Indexed: 04/11/2024]
Abstract
Modern imaging strategies are paramount to studying living systems such as cells, bacteria, and fungi and their response to pathogens, toxicants, and nanomaterials (NMs) as modulated by exposure and environmental factors. The need to understand the processes and mechanisms of damage, healing, and cell survivability of living systems continues to motivate the development of alternative imaging strategies. Of particular interest is the use of label-free techniques (microscopy procedures that do not require sample staining) that minimize interference of biological processes by foreign marking substances and reduce intense light exposure and potential photo-toxicity effects. This review focuses on the synergic capabilities of atomic force microscopy (AFM) as a well-developed and robust imaging strategy with demonstrated applications to unravel intimate details in biomedical applications, with the label-free, fast, and enduring Holotomographic Microscopy (HTM) strategy. HTM is a technique that combines holography and tomography using a low intensity continuous illumination laser to investigate (quantitatively and non-invasively) cells, microorganisms, and thin tissue by generating three-dimensional (3D) images and monitoring in real-time inner morphological changes. We first review the operating principles that form the basis for the complementary details provided by these techniques regarding the surface and internal information provided by HTM and AFM, which are essential and complimentary for the development of several biomedical areas studying the interaction mechanisms of NMs with living organisms. First, AFM can provide superb resolution on surface morphology and biomechanical characterization. Second, the quantitative phase capabilities of HTM enable superb modeling and quantification of the volume, surface area, protein content, and mass density of the main components of cells and microorganisms, including the morphology of cells in microbiological systems. These capabilities result from directly quantifying refractive index changes without requiring fluorescent markers or chemicals. As such, HTM is ideal for long-term monitoring of living organisms in conditions close to their natural settings. We present a case-based review of the principal uses of both techniques and their essential contributions to nanomedicine and nanotoxicology (study of the harmful effects of NMs in living organisms), emphasizing cancer and infectious disease control. The synergic impact of the sequential use of these complementary strategies provides a clear drive for adopting these techniques as interdependent fundamental tools.
Collapse
Affiliation(s)
- Iliana E Medina-Ramirez
- Department of Chemistry, Universidad Autónoma de Aguascalientes, Av. Universidad 940, Aguascalientes, Ags, Mexico.
| | - J E Macias-Diaz
- Department of Mathematics and Physics, Universidad Autónoma de Aguascalientes, Av. Universidad 940, Aguascalientes, Ags, Mexico
| | - David Masuoka-Ito
- Department of Stomatology, Universidad Autónoma de Aguascalientes, Av. Universidad 940, Aguascalientes, Ags, Mexico
| | - Juan Antonio Zapien
- Department of Materials Science and Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, People's Republic of China.
| |
Collapse
|
5
|
Di Santo R, Niccolini B, Romanò S, Vaccaro M, Di Giacinto F, De Spirito M, Ciasca G. Advancements in Mid-Infrared spectroscopy of extracellular vesicles. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 305:123346. [PMID: 37774583 DOI: 10.1016/j.saa.2023.123346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 08/07/2023] [Accepted: 09/02/2023] [Indexed: 10/01/2023]
Abstract
Extracellular vesicles (EVs) are lipid vesicles secreted by all cells into the extracellular space and act as nanosized biological messengers among cells. They carry a specific molecular cargo, composed of lipids, proteins, nucleic acids, and carbohydrates, which reflects the state of their parent cells. Due to their remarkable structural and compositional heterogeneity, characterizing EVs, particularly from a biochemical perspective, presents complex challenges. In this context, mid-infrared (IR) spectroscopy is emerging as a valuable tool, providing researchers with a comprehensive and label-free spectral fingerprint of EVs in terms of their specific molecular content. This review aims to provide an up-to-date critical overview of the major advancements in mid-IR spectroscopy of extracellular vesicles, encompassing both fundamental and applied research achievements. We also systematically emphasize the new possibilities offered by the integration of emerging cutting-edge IR technologies, such as tip-enhanced and surface-enhanced spectroscopy approaches, along with the growing use of machine learning for data analysis and spectral interpretation. Additionally, to assist researchers in navigating this intricate subject, our manuscript includes a wide and detailed collection of the spectral peaks that have been assigned to EV molecular constituents up to now in the literature.
Collapse
Affiliation(s)
- Riccardo Di Santo
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; Fondazione Policlinico Universitario "A. Gemelli" IRCCS, 00168 Rome, Italy.
| | - Benedetta Niccolini
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Sabrina Romanò
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Maria Vaccaro
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, 00168 Rome, Italy
| | - Flavio Di Giacinto
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; Fondazione Policlinico Universitario "A. Gemelli" IRCCS, 00168 Rome, Italy
| | - Marco De Spirito
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; Fondazione Policlinico Universitario "A. Gemelli" IRCCS, 00168 Rome, Italy
| | - Gabriele Ciasca
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; Fondazione Policlinico Universitario "A. Gemelli" IRCCS, 00168 Rome, Italy
| |
Collapse
|
6
|
Rayamajhi S, Sipes J, Tetlow AL, Saha S, Bansal A, Godwin AK. Extracellular Vesicles as Liquid Biopsy Biomarkers across the Cancer Journey: From Early Detection to Recurrence. Clin Chem 2024; 70:206-219. [PMID: 38175602 DOI: 10.1093/clinchem/hvad176] [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: 07/30/2023] [Accepted: 09/26/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Cancer is a dynamic process and thus requires highly informative and reliable biomarkers to help guide patient care. Liquid-based biopsies have emerged as a clinical tool for tracking cancer dynamics. Extracellular vesicles (EVs), lipid bilayer delimited particles secreted by cells, are a new class of liquid-based biomarkers. EVs are rich in selectively sorted biomolecule cargos, which provide a spatiotemporal fingerprint of the cell of origin, including cancer cells. CONTENT This review summarizes the performance characteristics of EV-based biomarkers at different stages of cancer progression, from early malignancy to recurrence, while emphasizing their potential as diagnostic, prognostic, and screening biomarkers. We discuss the characteristics of effective biomarkers, consider challenges associated with the EV biomarker field, and report guidelines based on the biomarker discovery pipeline. SUMMARY Basic science and clinical trial studies have shown the potential of EVs as precision-based biomarkers for tracking cancer status, with promising applications for diagnosing disease, predicting response to therapy, and tracking disease burden. The multi-analyte cargos of EVs enhance the performance characteristics of biomarkers. Recent technological advances in ultrasensitive detection of EVs have shown promise with high specificity and sensitivity to differentiate early-cancer cases vs healthy individuals, potentially outperforming current gold-standard imaging-based cancer diagnosis. Ultimately, clinical translation will be dictated by how these new EV biomarker-based platforms perform in larger sample cohorts. Applying ultrasensitive, scalable, and reproducible EV detection platforms with better design considerations based upon the biomarker discovery pipeline should guide the field towards clinically useful liquid biopsy biomarkers.
Collapse
Affiliation(s)
- Sagar Rayamajhi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States
| | - Jared Sipes
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States
| | - Ashley L Tetlow
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States
| | - Souvik Saha
- Division of Gastroenterology and Hepatology, University of Kansas Health System, Kansas City, KS, United States
| | - Ajay Bansal
- Division of Gastroenterology and Hepatology, University of Kansas Health System, Kansas City, KS, United States
- The University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, United States
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States
- The University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, United States
- Division of Genomic Diagnostics, University of Kansas Health System, Kansas City, KS, United States
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
7
|
Leggio L, Paternò G, Vivarelli S, Bonasera A, Pignataro B, Iraci N, Arrabito G. Label-free approaches for extracellular vesicle detection. iScience 2023; 26:108105. [PMID: 37867957 PMCID: PMC10589885 DOI: 10.1016/j.isci.2023.108105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023] Open
Abstract
Extracellular vesicles (EVs) represent pivotal mediators in cell-to-cell communication. They are lipid-membranous carriers of several biomolecules, which can be produced by almost all cells. In the current Era of precision medicine, EVs gained growing attention thanks to their potential in both biomarker discovery and nanotherapeutics applications. However, current technical limitations in isolating and/or detecting EVs restrain their standard use in clinics. This review explores all the state-of-the-art analytical technologies which are currently overcoming these issues. On one end, several innovative optical-, electrical-, and spectroscopy-based detection methods represent advantageous label-free methodologies for faster EV detection. On the other end, microfluidics-based lab-on-a-chip tools support EV purification from low-concentrated samples. Altogether, these technologies will strengthen the routine application of EVs in clinics.
Collapse
Affiliation(s)
- Loredana Leggio
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Greta Paternò
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Silvia Vivarelli
- Department of Biomedical and Dental Sciences, Morphological and Functional Imaging, Section of Occupational Medicine, University of Messina, Messina, Italy
| | - Aurelio Bonasera
- Department of Physics and Chemistry - Emilio Segrè, University of Palermo, Viale delle Scienze, building 17, 90128 Palermo, Italy
| | - Bruno Pignataro
- Department of Physics and Chemistry - Emilio Segrè, University of Palermo, Viale delle Scienze, building 17, 90128 Palermo, Italy
| | - Nunzio Iraci
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giuseppe Arrabito
- Department of Physics and Chemistry - Emilio Segrè, University of Palermo, Viale delle Scienze, building 17, 90128 Palermo, Italy
| |
Collapse
|
8
|
Xie X, Yu W, Chen Z, Wang L, Yang J, Liu S, Li L, Li Y, Huang Y. Early-stage oral cancer diagnosis by artificial intelligence-based SERS using Ag NWs@ZIF core-shell nanochains. NANOSCALE 2023; 15:13466-13472. [PMID: 37548371 DOI: 10.1039/d3nr02662k] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Surface-enhanced Raman spectroscopy (SERS) has great potential in the early diagnosis of diseases by detecting the changes of volatile biomarkers in exhaled breath, because of its high sensitivity, rich chemical molecular fingerprint information, and immunity to humidity. Here, an accurate diagnosis of oral cancer (OC) is demonstrated using artificial intelligence (AI)-based SERS of exhaled breath in plasmonic-metal organic framework (MOF) nanoparticles. These plasmonic-MOF nanoparticles were prepared using a zeolitic imidazolate framework coated on Ag nanowires (Ag NWs@ZIF), which offers Raman enhancement from the plasmonic nanowires and gas enrichment from the ZIF shells. Then, the core-shell nanochains of Ag NWs@ZIF prepared with 0.5 mL Ag NWs were selected to capture gaseous methanethiol, which is a tumor biomarker, from the exhalation of OC patients. The substrate was used to collect a total of 400 SERS spectra of exhaled breath of simulated healthy people and simulated OC patients. The artificial neural network (ANN) model in the AI algorithm was trained with these SERS spectra and could classify them with an accuracy of 99%. Notably, the model predicted OC with an area under the curve (AUC) of 0.996 for the simulated OC breath samples. This work suggests the great potential of the combination of breath analysis and AI as a method for the early-stage diagnosis of oral cancer.
Collapse
Affiliation(s)
- Xin Xie
- State Key Laboratory of Coal Mine Disaster Dynamics and Control, Chongqing Key Laboratory of Soft Condensed Matter Physics and Smart Materials, College of Physics, Chongqing University, Chongqing 400044, China.
| | - Wenrou Yu
- State Key Laboratory of Coal Mine Disaster Dynamics and Control, Chongqing Key Laboratory of Soft Condensed Matter Physics and Smart Materials, College of Physics, Chongqing University, Chongqing 400044, China.
| | - Zhaoxian Chen
- State Key Laboratory of Coal Mine Disaster Dynamics and Control, Chongqing Key Laboratory of Soft Condensed Matter Physics and Smart Materials, College of Physics, Chongqing University, Chongqing 400044, China.
| | - Li Wang
- School of Optoelectronics Engineering, Chongqing University, Chongqing 401331, China
| | - Junjun Yang
- State Key Laboratory of Coal Mine Disaster Dynamics and Control, Chongqing Key Laboratory of Soft Condensed Matter Physics and Smart Materials, College of Physics, Chongqing University, Chongqing 400044, China.
| | - Shihong Liu
- Department of Geriatric Oncology and Department of Palliative Care, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Linze Li
- State Key Laboratory of Coal Mine Disaster Dynamics and Control, Chongqing Key Laboratory of Soft Condensed Matter Physics and Smart Materials, College of Physics, Chongqing University, Chongqing 400044, China.
| | - Yanxi Li
- State Key Laboratory of Coal Mine Disaster Dynamics and Control, Chongqing Key Laboratory of Soft Condensed Matter Physics and Smart Materials, College of Physics, Chongqing University, Chongqing 400044, China.
| | - Yingzhou Huang
- State Key Laboratory of Coal Mine Disaster Dynamics and Control, Chongqing Key Laboratory of Soft Condensed Matter Physics and Smart Materials, College of Physics, Chongqing University, Chongqing 400044, China.
| |
Collapse
|
9
|
Mao Y, Liu P, Wei J, Xie Y, Zheng Q, Li R, Yao J. Cell Therapy for Androgenetic Alopecia: Elixir or Trick? Stem Cell Rev Rep 2023:10.1007/s12015-023-10532-2. [PMID: 37277541 PMCID: PMC10390634 DOI: 10.1007/s12015-023-10532-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2023] [Indexed: 06/07/2023]
Abstract
Androgenetic alopecia is the most common cause of hair loss aggravated by increased life pressure, tension, and anxiety. Although androgenetic alopecia (AGA) does not significantly effect physical health, it can have serious negative impact on the mental health and quality of life of the patient. Currently, the effect of medical treatment for AGA is not idealistic, stem cell-based regenerative medicine has shown potential for hair regrowth and follicle repair, but the long-term effect and mechanism of stem cell therapy is not quite explicit. In this review, we summarize the methods, efficacy, mechanism, and clinical progress of stem cell therapies for AGA by now, hope it will present a more comprehensive view in this topic.
Collapse
Affiliation(s)
- Yongcui Mao
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Pinyan Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Jiayun Wei
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Ye Xie
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Qiuxia Zheng
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Rui Li
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Jia Yao
- The First Clinical Medical College of Lanzhou University, Lanzhou, China.
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, China.
| |
Collapse
|
10
|
Shin H, Choi BH, Shim O, Kim J, Park Y, Cho SK, Kim HK, Choi Y. Single test-based diagnosis of multiple cancer types using Exosome-SERS-AI for early stage cancers. Nat Commun 2023; 14:1644. [PMID: 36964142 PMCID: PMC10039041 DOI: 10.1038/s41467-023-37403-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 03/16/2023] [Indexed: 03/26/2023] Open
Abstract
Early cancer detection has significant clinical value, but there remains no single method that can comprehensively identify multiple types of early-stage cancer. Here, we report the diagnostic accuracy of simultaneous detection of 6 types of early-stage cancers (lung, breast, colon, liver, pancreas, and stomach) by analyzing surface-enhanced Raman spectroscopy profiles of exosomes using artificial intelligence in a retrospective study design. It includes classification models that recognize signal patterns of plasma exosomes to identify both their presence and tissues of origin. Using 520 test samples, our system identified cancer presence with an area under the curve value of 0.970. Moreover, the system classified the tumor organ type of 278 early-stage cancer patients with a mean area under the curve of 0.945. The final integrated decision model showed a sensitivity of 90.2% at a specificity of 94.4% while predicting the tumor organ of 72% of positive patients. Since our method utilizes a non-specific analysis of Raman signatures, its diagnostic scope could potentially be expanded to include other diseases.
Collapse
Affiliation(s)
- Hyunku Shin
- EXoPERT Corporation, Seoul, 02580, Republic of Korea
| | - Byeong Hyeon Choi
- Department of Thoracic and Cardiovascular Surgery, College of Medicine, Korea University Guro Hospital, Seoul, 08308, Republic of Korea
- Korea Artificial Organ Center, Korea University, Seoul, 02841, Republic of Korea
| | - On Shim
- EXoPERT Corporation, Seoul, 02580, Republic of Korea
| | - Jihee Kim
- EXoPERT Corporation, Seoul, 02580, Republic of Korea
| | - Yong Park
- Division of Hematology-Oncology, Department of Internal Medicine, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Suk Ki Cho
- Division of Thoracic Surgery, Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea
| | - Hyun Koo Kim
- Department of Thoracic and Cardiovascular Surgery, College of Medicine, Korea University Guro Hospital, Seoul, 08308, Republic of Korea.
- Department of Biomedical Sciences, College of Medicine, Korea University, 02841, Seoul, Republic of Korea.
| | - Yeonho Choi
- EXoPERT Corporation, Seoul, 02580, Republic of Korea.
- School of Biomedical Engineering, Korea University, Seoul, 02841, Republic of Korea.
- Department of Biomedical Engineering, Korea University, Seoul, 02841, Republic of Korea.
- Interdisciplinary Program in Precision Public Health, Korea University, 02841, Seoul, Republic of Korea.
| |
Collapse
|
11
|
Extracellular Vesicles in Colorectal Cancer: From Tumor Growth and Metastasis to Biomarkers and Nanomedications. Cancers (Basel) 2023; 15:cancers15041107. [PMID: 36831450 PMCID: PMC9953945 DOI: 10.3390/cancers15041107] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Colorectal cancer (CRC) is a leading public health concern due to its incidence and high mortality rates, highlighting the requirement of an early diagnosis. Evaluation of circulating extracellular vesicles (EVs) might constitute a noninvasive and reliable approach for CRC detection and for patient follow-up because EVs display the molecular features of the cells they originate. EVs are released by almost all cell types and are mainly categorized as exosomes originating from exocytosis of intraluminal vesicles from multivesicular bodies, ectosomes resulting from outward budding of the plasma membrane and apoptotic bodies' ensuing cell shrinkage. These vesicles play a critical role in intercellular communications during physiological and pathological processes. They facilitate CRC progression and premetastatic niche formation, and they enable transfer of chemotherapy resistance to sensitive cells through the local or remote delivery of their lipid, nucleic acid and protein content. On another note, their stability in the bloodstream, their permeation in tissues and their sheltering of packaged material make engineered EVs suitable vectors for efficient delivery of tracers and therapeutic agents for tumor imaging or treatment. Here, we focus on the physiopathological role of EVs in CRCs, their value in the diagnosis and prognosis and ongoing investigations into therapeutic approaches.
Collapse
|
12
|
Eibl RH, Schneemann M. Medulloblastoma: From TP53 Mutations to Molecular Classification and Liquid Biopsy. BIOLOGY 2023; 12:267. [PMID: 36829544 PMCID: PMC9952923 DOI: 10.3390/biology12020267] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023]
Abstract
A recent paradigm shift in the diagnostics of medulloblastoma allowed the distinction of four major groups defined by genetic data rather than histology. This new molecular classification correlates better with prognosis and will allow for the better clinical management of therapies targeting druggable mutations, but also offer a new combination of monitoring tumor development in real-time and treatment response by sequential liquid biopsy. This review highlights recent developments after a century of milestones in neurosurgery and radio- and chemotherapy, but also controversial theories on the cell of origin, animal models, and the use of liquid biopsy.
Collapse
Affiliation(s)
- Robert H. Eibl
- c/o M. Schneemann; Department of Internal Medicine, Hospitals of Schaffhausen, 8208 Schaffhausen, Switzerland
| | - Markus Schneemann
- Department of Internal Medicine, Hospitals of Schaffhausen, 8208 Schaffhausen, Switzerland
| |
Collapse
|
13
|
Temperini ME, Di Giacinto F, Romanò S, Di Santo R, Augello A, Polito R, Baldassarre L, Giliberti V, Papi M, Basile U, Niccolini B, Krasnowska EK, Serafino A, De Spirito M, Di Gaspare A, Ortolani M, Ciasca G. Antenna-enhanced mid-infrared detection of extracellular vesicles derived from human cancer cell cultures. J Nanobiotechnology 2022; 20:530. [PMID: 36514065 PMCID: PMC9746222 DOI: 10.1186/s12951-022-01693-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 10/30/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Extracellular Vesicles (EVs) are sub-micrometer lipid-bound particles released by most cell types. They are considered a promising source of cancer biomarkers for liquid biopsy and personalized medicine due to their specific molecular cargo, which provides biochemical information on the state of parent cells. Despite this potential, EVs translation process in the diagnostic practice is still at its birth, and the development of novel medical devices for their detection and characterization is highly required. RESULTS In this study, we demonstrate mid-infrared plasmonic nanoantenna arrays designed to detect, in the liquid and dry phase, the specific vibrational absorption signal of EVs simultaneously with the unspecific refractive index sensing signal. For this purpose, EVs are immobilized on the gold nanoantenna surface by immunocapture, allowing us to select specific EV sub-populations and get rid of contaminants. A wet sample-handling technique relying on hydrophobicity contrast enables effortless reflectance measurements with a Fourier-transform infrared (FTIR) spectro-microscope in the wavelength range between 10 and 3 µm. In a proof-of-principle experiment carried out on EVs released from human colorectal adenocarcinoma (CRC) cells, the protein absorption bands (amide-I and amide-II between 5.9 and 6.4 µm) increase sharply within minutes when the EV solution is introduced in the fluidic chamber, indicating sensitivity to the EV proteins. A refractive index sensing curve is simultaneously provided by our sensor in the form of the redshift of a sharp spectral edge at wavelengths around 5 µm, where no vibrational absorption of organic molecules takes place: this permits to extract of the dynamics of EV capture by antibodies from the overall molecular layer deposition dynamics, which is typically measured by commercial surface plasmon resonance sensors. Additionally, the described metasurface is exploited to compare the spectral response of EVs derived from cancer cells with increasing invasiveness and metastatic potential, suggesting that the average secondary structure content in EVs can be correlated with cell malignancy. CONCLUSIONS Thanks to the high protein sensitivity and the possibility to work with small sample volumes-two key features for ultrasensitive detection of extracellular vesicles- our lab-on-chip can positively impact the development of novel laboratory medicine methods for the molecular characterization of EVs.
Collapse
Affiliation(s)
- Maria Eleonora Temperini
- grid.7841.aDepartment of Physics, Sapienza University of Rome, Piazzale Aldo Moro 2, 00185 Rome, Italy ,grid.25786.3e0000 0004 1764 2907Center for Life Neuro and Nano Sciences IIT@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy
| | - Flavio Di Giacinto
- grid.414603.4Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Rome, Italy ,grid.8142.f0000 0001 0941 3192Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Sabrina Romanò
- grid.414603.4Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Rome, Italy ,grid.8142.f0000 0001 0941 3192Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Riccardo Di Santo
- grid.414603.4Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Rome, Italy
| | - Alberto Augello
- grid.414603.4Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Rome, Italy
| | - Raffaella Polito
- grid.7841.aDepartment of Physics, Sapienza University of Rome, Piazzale Aldo Moro 2, 00185 Rome, Italy
| | - Leonetta Baldassarre
- grid.7841.aDepartment of Physics, Sapienza University of Rome, Piazzale Aldo Moro 2, 00185 Rome, Italy
| | - Valeria Giliberti
- grid.25786.3e0000 0004 1764 2907Center for Life Neuro and Nano Sciences IIT@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy
| | - Massimiliano Papi
- grid.414603.4Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Rome, Italy ,grid.8142.f0000 0001 0941 3192Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Umberto Basile
- grid.414603.4Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Benedetta Niccolini
- grid.8142.f0000 0001 0941 3192Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ewa K. Krasnowska
- grid.5326.20000 0001 1940 4177Institute of Translational Pharmacology, National Research Council of Italy, Rome, Italy
| | - Annalucia Serafino
- grid.5326.20000 0001 1940 4177Institute of Translational Pharmacology, National Research Council of Italy, Rome, Italy
| | - Marco De Spirito
- grid.414603.4Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Rome, Italy ,grid.8142.f0000 0001 0941 3192Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandra Di Gaspare
- grid.414603.4Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Rome, Italy ,grid.509494.5NEST, CNR-Istituto Nanoscienze and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Michele Ortolani
- grid.7841.aDepartment of Physics, Sapienza University of Rome, Piazzale Aldo Moro 2, 00185 Rome, Italy ,grid.25786.3e0000 0004 1764 2907Center for Life Neuro and Nano Sciences IIT@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy
| | - Gabriele Ciasca
- grid.414603.4Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Rome, Italy ,grid.8142.f0000 0001 0941 3192Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
14
|
Cansever Mutlu E, Kaya M, Küçük I, Ben-Nissan B, Stamboulis A. Exosome Structures Supported by Machine Learning Can Be Used as a Promising Diagnostic Tool. MATERIALS (BASEL, SWITZERLAND) 2022; 15:7967. [PMID: 36431454 PMCID: PMC9693854 DOI: 10.3390/ma15227967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/23/2022] [Accepted: 11/02/2022] [Indexed: 06/16/2023]
Abstract
Principal component analysis (PCA) as a machine-learning technique could serve in disease diagnosis and prognosis by evaluating the dynamic morphological features of exosomes via Cryo-TEM-imaging. This hypothesis was investigated after the crude isolation of similarly featured exosomes derived from the extracellular vehicles (EVs) of immature dendritic cells (IDCs) JAWSII. It is possible to identify functional molecular groups by FTIR, but the unique physical and morphological characteristics of exosomes can only be revealed by specialized imaging techniques such as cryo-TEM. On the other hand, PCA has the ability to examine the morphological features of each of these IDC-derived exosomes by considering software parameters such as various membrane projections and differences in Gaussians, Hessian, hue, and class to assess the 3D orientation, shape, size, and brightness of the isolated IDC-derived exosome structures. In addition, Brownian motions from nanoparticle tracking analysis of EV IDC-derived exosomes were also compared with EV IDC-derived exosome images collected by scanning electron microscopy and confocal microscopy. Sodium-Dodecyl-Sulphate-Polyacrylamide-Gel-Electrophoresis (SDS-PAGE) was performed to separate the protein content of the crude isolates showing that no considerable protein contamination occurred during the crude isolation technique of IDC-derived-exosomes. This is an important finding because no additional purification of these exosomes is required, making PCA analysis both valuable and novel.
Collapse
Affiliation(s)
- Esra Cansever Mutlu
- College of Engineering and Physical Science, School of Metallurgy and Materials, Biomaterials Research Group, University of Birmingham, Birmingham B15 2TT, UK
- Department of Biomedical Engineering, Faculty of Engineering and Architecture, Beykent University, Sarıyer, 34398 İstanbul, Türkiye
| | - Mustafa Kaya
- Department of Biomedical Engineering, Faculty of Engineering and Architecture, Beykent University, Sarıyer, 34398 İstanbul, Türkiye
- Institute of Nanotechnology, Gebze Technical University, 41400 Gebze, Türkiye
| | - Israfil Küçük
- Institute of Nanotechnology, Gebze Technical University, 41400 Gebze, Türkiye
| | - Besim Ben-Nissan
- School of Life Sciences, Translational Biomaterials and Medicine Group, University of Technology Sydney, P.O. Box 123, Broadway, NSW 2007, Australia
| | - Artemis Stamboulis
- College of Engineering and Physical Science, School of Metallurgy and Materials, Biomaterials Research Group, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
15
|
Nardini M, Ciasca G, Lauria A, Rossi C, Di Giacinto F, Romanò S, Di Santo R, Papi M, Palmieri V, Perini G, Basile U, Alcaro FD, Di Stasio E, Bizzarro A, Masullo C, De Spirito M. Sensing red blood cell nano-mechanics: Toward a novel blood biomarker for Alzheimer's disease. Front Aging Neurosci 2022; 14:932354. [PMID: 36204549 PMCID: PMC9530048 DOI: 10.3389/fnagi.2022.932354] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Red blood cells (RBCs) are characterized by a remarkable elasticity, which allows them to undergo very large deformation when passing through small vessels and capillaries. This extreme deformability is altered in various clinical conditions, suggesting that the analysis of red blood cell (RBC) mechanics has potential applications in the search for non-invasive and cost-effective blood biomarkers. Here, we provide a comparative study of the mechanical response of RBCs in patients with Alzheimer's disease (AD) and healthy subjects. For this purpose, RBC viscoelastic response was investigated using atomic force microscopy (AFM) in the force spectroscopy mode. Two types of analyses were performed: (i) a conventional analysis of AFM force-distance (FD) curves, which allowed us to retrieve the apparent Young's modulus, E; and (ii) a more in-depth analysis of time-dependent relaxation curves in the framework of the standard linear solid (SLS) model, which allowed us to estimate cell viscosity and elasticity, independently. Our data demonstrate that, while conventional analysis of AFM FD curves fails in distinguishing the two groups, the mechanical parameters obtained with the SLS model show a very good classification ability. The diagnostic performance of mechanical parameters was assessed using receiving operator characteristic (ROC) curves, showing very large areas under the curves (AUC) for selected biomarkers (AUC > 0.9). Taken all together, the data presented here demonstrate that RBC mechanics are significantly altered in AD, also highlighting the key role played by viscous forces. These RBC abnormalities in AD, which include both a modified elasticity and viscosity, could be considered a potential source of plasmatic biomarkers in the field of liquid biopsy to be used in combination with more established indicators of the pathology.
Collapse
Affiliation(s)
- Matteo Nardini
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Gabriele Ciasca
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Alessandra Lauria
- Unitá Operativa Complessa Neuroriabilitazione ad Alta Intensitá, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Cristina Rossi
- Department of Laboratory Diagnostic and Infectious Diseases, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Flavio Di Giacinto
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Sabrina Romanò
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Riccardo Di Santo
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Massimiliano Papi
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Valentina Palmieri
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
- Istituto dei Sistemi Complessi (ISC), Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Giordano Perini
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Umberto Basile
- Department of Laboratory Diagnostic and Infectious Diseases, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Francesca D. Alcaro
- Department of Laboratory Diagnostic and Infectious Diseases, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Enrico Di Stasio
- Department of Laboratory Diagnostic and Infectious Diseases, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Alessandra Bizzarro
- Unitáă Operativa Complessa Continuità assistenziale, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Carlo Masullo
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
- Sezione di Neurologia, Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marco De Spirito
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
16
|
Han Z, Peng X, Yang Y, Yi J, Zhao D, Bao Q, Long S, Yu SX, Xu XX, Liu B, Liu YJ, Shen Y, Qiao L. Integrated microfluidic-SERS for exosome biomarker profiling and osteosarcoma diagnosis. Biosens Bioelectron 2022; 217:114709. [PMID: 36115123 DOI: 10.1016/j.bios.2022.114709] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/06/2022] [Indexed: 12/18/2022]
Abstract
Osteosarcoma is one of the most frequent primary sarcoma of bone among adolescents. Early diagnosis of osteosarcoma is the key factor to achieve high survival rate of patients. Nevertheless, traditional histological biopsy is highly invasive and associated with the risk of arousing tumor spread. Herein, we develop a method integrating microfluidics and surface-enhanced Raman spectroscopy (SERS) to isolate plasma-derived exosomes and profile multiple exosomal biomarkers for the diagnosis of osteosarcoma. The method showed highly efficient isolation of exosomes directly from human plasma and can profile exosomes based on protein biomarkers, with the detection limit down to 2 exosomes per μL. The whole assay can be performed in 5 h and only consumed 50 μL of plasma for one analysis. With the method, we analyzed the level of three protein biomarkers, i.e., CD63, vimentin (VIM) and epithelial cell adhesion molecule (EpCAM), on plasma-derived exosomes from 20 osteosarcoma patients and 20 heathy controls. Significantly higher levels of CD63, VIM and EpCAM were observed on plasma exosomes from the osteosarcoma patients compared to the healthy controls. Based on the level of the exosomal biomarkers, a classification model was built for the rapid diagnosis of osteosarcoma, with the sensitivity, specificity and accuracy of 100%, 90% and 95%, respectively. The proposed method does not require complex operations nor expensive equipment, and has great promise in clinical diagnosis of cancer as a liquid biopsy technique.
Collapse
Affiliation(s)
- Zhenzhen Han
- Department of Chemistry and Shanghai Stomatological Hospital, Fudan University, Shanghai, 200000, China; Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Xinyan Peng
- Department of Chemistry and Shanghai Stomatological Hospital, Fudan University, Shanghai, 200000, China
| | - Yi Yang
- Department of Chemistry and Shanghai Stomatological Hospital, Fudan University, Shanghai, 200000, China
| | - Jia Yi
- Department of Chemistry and Shanghai Stomatological Hospital, Fudan University, Shanghai, 200000, China
| | - Dan Zhao
- Department of Chemistry and Shanghai Stomatological Hospital, Fudan University, Shanghai, 200000, China
| | - Qiyuan Bao
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200000, China
| | - Shuping Long
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Sai-Xi Yu
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Department of Systems Biology for Medicine, Fudan University, China
| | - Xin-Xin Xu
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Department of Systems Biology for Medicine, Fudan University, China
| | - Baohong Liu
- Department of Chemistry and Shanghai Stomatological Hospital, Fudan University, Shanghai, 200000, China
| | - Yan-Jun Liu
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Department of Systems Biology for Medicine, Fudan University, China
| | - Yuhui Shen
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200000, China
| | - Liang Qiao
- Department of Chemistry and Shanghai Stomatological Hospital, Fudan University, Shanghai, 200000, China.
| |
Collapse
|
17
|
Molecular Recognition of Surface Trans-Sialidases in Extracellular Vesicles of the Parasite Trypanosoma cruzi Using Atomic Force Microscopy (AFM). Int J Mol Sci 2022; 23:ijms23137193. [PMID: 35806197 PMCID: PMC9266976 DOI: 10.3390/ijms23137193] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/23/2022] [Accepted: 06/26/2022] [Indexed: 02/06/2023] Open
Abstract
Trans-sialidases (TS) are important constitutive macromolecules of the secretome present on the surface of Trypanosoma cruzi (T. cruzi) that play a central role as a virulence factor in Chagas disease. These enzymes have been related to infectivity, escape from immune surveillance and pathogenesis exhibited by this protozoan parasite. In this work, atomic force microscopy (AFM)-based single molecule-force spectroscopy is implemented as a suitable technique for the detection and location of functional TS on the surface of extracellular vesicles (EVs) released by tissue-culture cell-derived trypomastigotes (Ex-TcT). For that purpose, AFM cantilevers with functionalized tips bearing the anti-TS monoclonal antibody mAb 39 as a sense biomolecule are engineered using a covalent chemical ligation based on vinyl sulfonate click chemistry; a reliable, simple and efficient methodology for the molecular recognition of TS using the antibody-antigen interaction. Measurements of the breakdown forces between anti-TS mAb 39 antibodies and EVs performed to elucidate adhesion and forces involved in the recognition events demonstrate that EVs isolated from tissue-culture cell-derived trypomastigotes of T. cruzi are enriched in TS. Additionally, a mapping of the TS binding sites with submicrometer-scale resolution is provided. This work represents the first AFM-based molecular recognition study of Ex-TcT using an antibody-tethered AFM probe.
Collapse
|
18
|
Mousavi SM, Amin Mahdian SM, Ebrahimi MS, Taghizadieh M, Vosough M, Sadri Nahand J, Hosseindoost S, Vousooghi N, Javar HA, Larijani B, Hadjighassem MR, Rahimian N, Hamblin MR, Mirzaei H. Microfluidics for detection of exosomes and microRNAs in cancer: State of the art. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 28:758-791. [PMID: 35664698 PMCID: PMC9130092 DOI: 10.1016/j.omtn.2022.04.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Exosomes are small extracellular vesicles with sizes ranging from 30-150 nanometers that contain proteins, lipids, mRNAs, microRNAs, and double-stranded DNA derived from the cells of origin. Exosomes can be taken up by target cells, acting as a means of cell-to-cell communication. The discovery of these vesicles in body fluids and their participation in cell communication has led to major breakthroughs in diagnosis, prognosis, and treatment of several conditions (e.g., cancer). However, conventional isolation and evaluation of exosomes and their microRNA content suffers from high cost, lengthy processes, difficult standardization, low purity, and poor yield. The emergence of microfluidics devices with increased efficiency in sieving, trapping, and immunological separation of small volumes could provide improved detection and monitoring of exosomes involved in cancer. Microfluidics techniques hold promise for advances in development of diagnostic and prognostic devices. This review covers ongoing research on microfluidics devices for detection of microRNAs and exosomes as biomarkers and their translation to point-of-care and clinical applications.
Collapse
Affiliation(s)
- Seyed Mojtaba Mousavi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Amin Mahdian
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Saeid Ebrahimi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Taghizadieh
- Department of Pathology, School of Medicine, Center for Women’s Health Research Zahra, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran
| | - Javid Sadri Nahand
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saereh Hosseindoost
- Pain Research Center, Neuroscience Institute, Tehran University of Medical Science, Tehran, Iran
| | - Nasim Vousooghi
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Cognitive and Behavioral Sciences, Tehran University of Medical Sciences, Tehran, Iran
- Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Akbari Javar
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Reza Hadjighassem
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Brain and Spinal Cord Research Center, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
19
|
Di Santo R, Vaccaro M, Romanò S, Di Giacinto F, Papi M, Rapaccini GL, De Spirito M, Miele L, Basile U, Ciasca G. Machine Learning-Assisted FTIR Analysis of Circulating Extracellular Vesicles for Cancer Liquid Biopsy. J Pers Med 2022; 12:jpm12060949. [PMID: 35743734 PMCID: PMC9224706 DOI: 10.3390/jpm12060949] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/01/2022] [Accepted: 06/05/2022] [Indexed: 12/20/2022] Open
Abstract
Extracellular vesicles (EVs) are abundantly released into the systemic circulation, where they show remarkable stability and harbor molecular constituents that provide biochemical information about their cells of origin. Due to this characteristic, EVs are attracting increasing attention as a source of circulating biomarkers for cancer liquid biopsy and personalized medicine. Despite this potential, none of the discovered biomarkers has entered the clinical practice so far, and novel approaches for the label-free characterization of EVs are highly demanded. In this regard, Fourier Transform Infrared Spectroscopy (FTIR) has great potential as it provides a quick, reproducible, and informative biomolecular fingerprint of EVs. In this pilot study, we investigated, for the first time in the literature, the capability of FTIR spectroscopy to distinguish between EVs extracted from sera of cancer patients and controls based on their mid-IR spectral response. For this purpose, EV-enriched suspensions were obtained from the serum of patients diagnosed with Hepatocellular Carcinoma (HCC) of nonviral origin and noncancer subjects. Our data point out the presence of statistically significant differences in the integrated intensities of major mid-IR absorption bands, including the carbohydrate and nucleic acids band, the protein amide I and II bands, and the lipid CH stretching band. Additionally, we used Principal Component Analysis combined with Linear Discriminant Analysis (PCA-LDA) for the automated classification of spectral data according to the shape of specific mid-IR spectral signatures. The diagnostic performances of the proposed spectral biomarkers, alone and combined, were evaluated using multivariate logistic regression followed by a Receiving Operator Curve analysis, obtaining large Areas Under the Curve (AUC = 0.91, 95% CI 0.81–1.0). Very interestingly, our analyses suggest that the discussed spectral biomarkers can outperform the classification ability of two widely used circulating HCC markers measured on the same groups of subjects, namely alpha-fetoprotein (AFP), and protein induced by the absence of vitamin K or antagonist-II (PIVKA-II).
Collapse
Affiliation(s)
- Riccardo Di Santo
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (M.V.); (F.D.G.); (M.P.); (G.L.R.); (M.D.S.); (G.C.)
- Correspondence:
| | - Maria Vaccaro
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (M.V.); (F.D.G.); (M.P.); (G.L.R.); (M.D.S.); (G.C.)
| | - Sabrina Romanò
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Flavio Di Giacinto
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (M.V.); (F.D.G.); (M.P.); (G.L.R.); (M.D.S.); (G.C.)
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Massimiliano Papi
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (M.V.); (F.D.G.); (M.P.); (G.L.R.); (M.D.S.); (G.C.)
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Gian Ludovico Rapaccini
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (M.V.); (F.D.G.); (M.P.); (G.L.R.); (M.D.S.); (G.C.)
- Sezione di Medicina Interna, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Marco De Spirito
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (M.V.); (F.D.G.); (M.P.); (G.L.R.); (M.D.S.); (G.C.)
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Luca Miele
- Sezione di Medicina Interna, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Umberto Basile
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy;
| | - Gabriele Ciasca
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (M.V.); (F.D.G.); (M.P.); (G.L.R.); (M.D.S.); (G.C.)
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| |
Collapse
|
20
|
Bragina VA, Khomyakova E, Orlov AV, Znoyko SL, Mochalova EN, Paniushkina L, Shender VO, Erbes T, Evtushenko EG, Bagrov DV, Lavrenova VN, Nazarenko I, Nikitin PI. Highly Sensitive Nanomagnetic Quantification of Extracellular Vesicles by Immunochromatographic Strips: A Tool for Liquid Biopsy. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:1579. [PMID: 35564289 PMCID: PMC9101557 DOI: 10.3390/nano12091579] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/18/2022] [Accepted: 05/02/2022] [Indexed: 01/27/2023]
Abstract
Extracellular vesicles (EVs) are promising agents for liquid biopsy-a non-invasive approach for the diagnosis of cancer and evaluation of therapy response. However, EV potential is limited by the lack of sufficiently sensitive, time-, and cost-efficient methods for their registration. This research aimed at developing a highly sensitive and easy-to-use immunochromatographic tool based on magnetic nanoparticles for EV quantification. The tool is demonstrated by detection of EVs isolated from cell culture supernatants and various body fluids using characteristic biomarkers, CD9 and CD81, and a tumor-associated marker-epithelial cell adhesion molecules. The detection limit of 3.7 × 105 EV/µL is one to two orders better than the most sensitive traditional lateral flow system and commercial ELISA kits. The detection specificity is ensured by an isotype control line on the test strip. The tool's advantages are due to the spatial quantification of EV-bound magnetic nanolabels within the strip volume by an original electronic technique. The inexpensive tool, promising for liquid biopsy in daily clinical routines, can be extended to other relevant biomarkers.
Collapse
Affiliation(s)
- Vera A. Bragina
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov St., 119991 Moscow, Russia; (V.A.B.); (E.K.); (A.V.O.); (S.L.Z.); (E.N.M.)
| | - Elena Khomyakova
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov St., 119991 Moscow, Russia; (V.A.B.); (E.K.); (A.V.O.); (S.L.Z.); (E.N.M.)
| | - Alexey V. Orlov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov St., 119991 Moscow, Russia; (V.A.B.); (E.K.); (A.V.O.); (S.L.Z.); (E.N.M.)
- Moscow Institute of Physics and Technology, 9 Institutskii per., 141700 Dolgoprudny, Russia
| | - Sergey L. Znoyko
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov St., 119991 Moscow, Russia; (V.A.B.); (E.K.); (A.V.O.); (S.L.Z.); (E.N.M.)
| | - Elizaveta N. Mochalova
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov St., 119991 Moscow, Russia; (V.A.B.); (E.K.); (A.V.O.); (S.L.Z.); (E.N.M.)
- Sirius University of Science and Technology, 1 Olympic Ave., 354340 Sochi, Russia
| | - Liliia Paniushkina
- Institute for Infection Prevention and Hospital Epidemiology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (L.P.); (I.N.)
| | - Victoria O. Shender
- Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, 1a Malaya Pirogovskaya St., 119992 Moscow, Russia; (V.O.S.); (V.N.L.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., 117997 Moscow, Russia
| | - Thalia Erbes
- Department of Obstetrics and Gynecology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany;
| | - Evgeniy G. Evtushenko
- Lomonosov Moscow State University, 1 Leninskie Gory, 119991 Moscow, Russia; (E.G.E.); (D.V.B.)
| | - Dmitry V. Bagrov
- Lomonosov Moscow State University, 1 Leninskie Gory, 119991 Moscow, Russia; (E.G.E.); (D.V.B.)
| | - Victoria N. Lavrenova
- Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, 1a Malaya Pirogovskaya St., 119992 Moscow, Russia; (V.O.S.); (V.N.L.)
- Lomonosov Moscow State University, 1 Leninskie Gory, 119991 Moscow, Russia; (E.G.E.); (D.V.B.)
| | - Irina Nazarenko
- Institute for Infection Prevention and Hospital Epidemiology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (L.P.); (I.N.)
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Petr I. Nikitin
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov St., 119991 Moscow, Russia; (V.A.B.); (E.K.); (A.V.O.); (S.L.Z.); (E.N.M.)
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), 31 Kashirskoe Shosse, 115409 Moscow, Russia
| |
Collapse
|
21
|
Liu T, Li Z, Li X, Zhao R, Wei X, Wang Z, Xin SX. In vivo visualization of murine melanoma cells B16-derived exosomes through magnetic resonance imaging. Biochim Biophys Acta Gen Subj 2022; 1866:130062. [PMID: 34822924 DOI: 10.1016/j.bbagen.2021.130062] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 10/22/2021] [Accepted: 11/17/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Numerous studies demonstrated that exosomes play a powerful role in mediating intercellular communication to induce a pro-tumoral environment to promote tumor progression, including pre-metastatic niche formation and metastasis. Noninvasive imaging could determine the in vivo kinetics of exosomes in real time to provide better understanding of the mechanisms of the tumor formation, progression and metastasis. Magnetic resonance imaging (MRI) is an ideal technique which provides excellent anatomical resolution, intrinsic soft tissue contrast, unlimited penetration depth and no radiation exposure. METHODS A fusion protein composed of ferritin heavy chain (FTH1) and lactadherin was designed for visualizing exosomes through MRI. FTH1 was served as MRI reporter protein and lactadherin is a membrane-associated protein that is distributed on exosome surface. The characterizations of labeled exosomes were validated through transmission electron microscopy, western blot, nanoparticle tracking analysis and finally visualized in vitro and in vivo through MRI. RESULTS MR imaging showed that the labeled exosomes are able to be visualized in vitro and in vivo. Verification of the characterizations of exosomes observed no significant difference between labeled and unlabeled exosomes. CONCLUSION The proposed FTH1 labeling method was useful for visualizing exosomes through MRI. GENERAL SIGNIFICANCE The present study first reported a novel self-label method for imaging labeled exosomes of tumor cells in vivo through MR with cell endogenous MRI reporter protein. It may be further used as a tool to enhance understanding the role of exosomes in various pathophysiological conditions.
Collapse
Affiliation(s)
- Tianqi Liu
- School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Zhenlin Li
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Xiaodong Li
- School of Medicine, South China University of Technology, Guangzhou 510006, Guangdong, China
| | - Ruiting Zhao
- School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Xinhua Wei
- Department of Radiology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, Guangdong, China
| | - Zixin Wang
- School of Electronics and Information Technology, Sun Yat-Sen University, Xingang Xi Road 135, Guangzhou 510275, Guangdong, China
| | - Sherman Xuegang Xin
- School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, Guangdong, China; School of Medicine, South China University of Technology, Guangzhou 510006, Guangdong, China.
| |
Collapse
|
22
|
Romanò S, Di Giacinto F, Primiano A, Gervasoni J, Mazzini A, Papi M, Urbani A, Serafino A, De Spirito M, Krasnowska EK, Ciasca G. Label-free spectroscopic characterization of exosomes reveals cancer cell differentiation. Anal Chim Acta 2022; 1192:339359. [DOI: 10.1016/j.aca.2021.339359] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 11/05/2021] [Accepted: 11/06/2021] [Indexed: 12/11/2022]
|
23
|
Ramirez-Garrastacho M, Bajo-Santos C, Line A, Martens-Uzunova ES, de la Fuente JM, Moros M, Soekmadji C, Tasken KA, Llorente A. Extracellular vesicles as a source of prostate cancer biomarkers in liquid biopsies: a decade of research. Br J Cancer 2022; 126:331-350. [PMID: 34811504 PMCID: PMC8810769 DOI: 10.1038/s41416-021-01610-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 01/02/2023] Open
Abstract
Prostate cancer is a global cancer burden and considerable effort has been made through the years to identify biomarkers for the disease. Approximately a decade ago, the potential of analysing extracellular vesicles in liquid biopsies started to be envisaged. This was the beginning of a new exciting area of research investigating the rich molecular treasure found in extracellular vesicles to identify biomarkers for a variety of diseases. Vesicles released from prostate cancer cells and cells of the tumour microenvironment carry molecular information about the disease that can be analysed in several biological fluids. Numerous studies document the interest of researchers in this field of research. However, methodological issues such as the isolation of vesicles have been challenging. Remarkably, novel technologies, including those based on nanotechnology, show promise for the further development and clinical use of extracellular vesicles as liquid biomarkers. Development of biomarkers is a long and complicated process, and there are still not many biomarkers based on extracellular vesicles in clinical use. However, the knowledge acquired during the last decade constitutes a solid basis for the future development of liquid biopsy tests for prostate cancer. These are urgently needed to bring prostate cancer treatment to the next level in precision medicine.
Collapse
Affiliation(s)
- Manuel Ramirez-Garrastacho
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | | | - Aija Line
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Elena S Martens-Uzunova
- Erasmus MC Cancer Institute, University Medical Center Rotterdam, Department of Urology, Laboratory of Experimental Urology, Erasmus MC, Rotterdam, The Netherlands
| | - Jesus Martinez de la Fuente
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Maria Moros
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Carolina Soekmadji
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Kristin Austlid Tasken
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Alicia Llorente
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
- Department for Mechanical, Electronics and Chemical Engineering, Oslo Metropolitan University, Oslo, Norway.
| |
Collapse
|
24
|
Fourier Transform Infrared (FTIR) Spectroscopy to Analyse Human Blood over the Last 20 Years: A Review towards Lab-on-a-Chip Devices. MICROMACHINES 2022; 13:mi13020187. [PMID: 35208311 PMCID: PMC8879834 DOI: 10.3390/mi13020187] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 01/27/2023]
Abstract
Since microorganisms are evolving rapidly, there is a growing need for a new, fast, and precise technique to analyse blood samples and distinguish healthy from pathological samples. Fourier Transform Infrared (FTIR) spectroscopy can provide information related to the biochemical composition and how it changes when a pathological state arises. FTIR spectroscopy has undergone rapid development over the last decades with a promise of easier, faster, and more impartial diagnoses within the biomedical field. However, thus far only a limited number of studies have addressed the use of FTIR spectroscopy in this field. This paper describes the main concepts related to FTIR and presents the latest research focusing on FTIR spectroscopy technology and its integration in lab-on-a-chip devices and their applications in the biological field. This review presents the potential use of FTIR to distinguish between healthy and pathological samples, with examples of early cancer detection, human immunodeficiency virus (HIV) detection, and routine blood analysis, among others. Finally, the study also reflects on the features of FTIR technology that can be applied in a lab-on-a-chip format and further developed for small healthcare devices that can be used for point-of-care monitoring purposes. To the best of the authors’ knowledge, no other published study has reviewed these topics. Therefore, this analysis and its results will fill this research gap.
Collapse
|
25
|
Ridolfi A, Caselli L, Baldoni M, Montis C, Mercuri F, Berti D, Valle F, Brucale M. Stiffness of Fluid and Gel Phase Lipid Nanovesicles: Weighting the Contributions of Membrane Bending Modulus and Luminal Pressurization. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:12027-12037. [PMID: 34610740 DOI: 10.1021/acs.langmuir.1c01660] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
The mechanical properties of biogenic membranous compartments are thought to be relevant in numerous biological processes; however, their quantitative measurement remains challenging for most of the already available force spectroscopy (FS)-based techniques. In particular, the debate on the mechanics of lipid nanovesicles and on the interpretation of their mechanical response to an applied force is still open. This is mostly due to the current lack of a unified model being able to describe the mechanical response of both gel and fluid phase lipid vesicles and to disentangle the contributions of membrane rigidity and luminal pressure. In this framework, we herein propose a simple model in which the interplay of membrane rigidity and luminal pressure to the overall vesicle stiffness is described as a series of springs; this approach allows estimating these two contributions for both gel and fluid phase liposomes. Atomic force microscopy-based FS, performed on both vesicles and supported lipid bilayers, is exploited for obtaining all the parameters involved in the model. Moreover, the use of coarse-grained full-scale molecular dynamics simulations allowed for better understanding of the differences in the mechanical responses of gel and fluid phase bilayers and supported the experimental findings. The results suggest that the pressure contribution is similar among all the probed vesicle types; however, it plays a dominant role in the mechanical response of lipid nanovesicles presenting a fluid phase membrane, while its contribution becomes comparable to the one of membrane rigidity in nanovesicles with a gel phase lipid membrane. The results presented herein offer a simple way to quantify two of the most important parameters in vesicle nanomechanics (membrane rigidity and internal pressurization), and as such represent a first step toward a currently unavailable, unified model for the mechanical response of gel and fluid phase lipid nanovesicles.
Collapse
Affiliation(s)
- Andrea Ridolfi
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande Interfase, 50019 Firenze, Italy
- Istituto per lo Studio dei Materiali Nanostrutturati, Consiglio Nazionale delle Ricerche, 40129 Bologna, Italy
- Dipartimento di Chimica "Ugo Schiff", Università degli Studi di Firenze, 50019 Firenze, Italy
| | - Lucrezia Caselli
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande Interfase, 50019 Firenze, Italy
- Dipartimento di Chimica "Ugo Schiff", Università degli Studi di Firenze, 50019 Firenze, Italy
| | - Matteo Baldoni
- Istituto per lo Studio dei Materiali Nanostrutturati, Consiglio Nazionale delle Ricerche, 40129 Bologna, Italy
| | - Costanza Montis
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande Interfase, 50019 Firenze, Italy
- Dipartimento di Chimica "Ugo Schiff", Università degli Studi di Firenze, 50019 Firenze, Italy
| | - Francesco Mercuri
- Istituto per lo Studio dei Materiali Nanostrutturati, Consiglio Nazionale delle Ricerche, 40129 Bologna, Italy
| | - Debora Berti
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande Interfase, 50019 Firenze, Italy
- Dipartimento di Chimica "Ugo Schiff", Università degli Studi di Firenze, 50019 Firenze, Italy
| | - Francesco Valle
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande Interfase, 50019 Firenze, Italy
- Istituto per lo Studio dei Materiali Nanostrutturati, Consiglio Nazionale delle Ricerche, 40129 Bologna, Italy
| | - Marco Brucale
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande Interfase, 50019 Firenze, Italy
- Istituto per lo Studio dei Materiali Nanostrutturati, Consiglio Nazionale delle Ricerche, 40129 Bologna, Italy
| |
Collapse
|
26
|
Bortot B, Apollonio M, Rampazzo E, Valle F, Brucale M, Ridolfi A, Ura B, Addobbati R, Di Lorenzo G, Romano F, Buonomo F, Ripepi C, Ricci G, Biffi S. Small extracellular vesicles from malignant ascites of patients with advanced ovarian cancer provide insights into the dynamics of the extracellular matrix. Mol Oncol 2021; 15:3596-3614. [PMID: 34614287 PMCID: PMC8637559 DOI: 10.1002/1878-0261.13110] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/31/2021] [Accepted: 10/04/2021] [Indexed: 12/30/2022] Open
Abstract
The exact role of malignant ascites in the development of intraperitoneal metastases remains unclear, and the mechanisms by which extracellular vesicles (EVs) promote tumor progression in the pre-metastatic niche have not been fully discovered. In this study, we characterized ascites from high-grade epithelial ovarian cancer patients. Small-EVs (30-150 nm) were isolated from two sources-the bulk ascites and the ascitic fluid-derived tumor cell cultures-and assessed with a combination of imaging, proteomic profiling, and protein expression analyses. In addition, Gene Ontology and pathway analysis were performed using different databases and bioinformatic tools. The results proved that the small-EVs derived from the two sources exhibited significantly different stiffness and size distributions. The bulk ascitic fluid-derived small-EVs were predominantly involved in the complement and coagulation cascade. Small-EVs derived from ascites cell cultures contained a robust proteomic profile of extracellular matrix remodeling regulators, and we observed an increase in transforming growth factor-β-I (TGFβI), plasminogen activator inhibitor 1 (PAI-1), and fibronectin expression after neoadjuvant chemotherapy. When measured in the two sources, we demonstrated that fibronectin exhibited opposite expression patterns in small-EVs in response to chemotherapy. These findings highlight the importance of an ascites cell isolation workflow in investigating the treatment-induced cancer adaption processes.
Collapse
Affiliation(s)
- Barbara Bortot
- Department of Medical Genetics, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Maura Apollonio
- Pediatric Department, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Enrico Rampazzo
- Department of Chemistry "Giacomo Ciamician", University of Bologna, Italy
| | - Francesco Valle
- Consorzio Sistemi a Grande Interfase, Department of Chemistry, University of Firenze, Italy.,Consiglio Nazionale delle Ricerche, Istituto per lo Studio dei Materiali Nanostrutturati (CNRISMN), Bologna, Italy
| | - Marco Brucale
- Consorzio Sistemi a Grande Interfase, Department of Chemistry, University of Firenze, Italy.,Consiglio Nazionale delle Ricerche, Istituto per lo Studio dei Materiali Nanostrutturati (CNRISMN), Bologna, Italy
| | - Andrea Ridolfi
- Consorzio Sistemi a Grande Interfase, Department of Chemistry, University of Firenze, Italy.,Department of Chemistry, University of Firenze, Italy
| | - Blendi Ura
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Riccardo Addobbati
- Department of Clinical Toxicology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Giovanni Di Lorenzo
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Federico Romano
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Francesca Buonomo
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Chiara Ripepi
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Giuseppe Ricci
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy.,Clinical Department of Medical, Surgical and Health Sciences, University of Trieste, Italy
| | - Stefania Biffi
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| |
Collapse
|