1
|
Shrivastava A, Kumar A, Aggarwal LM, Pradhan S, Choudhary S, Ashish A, Kashyap K, Mishra S. Evolution of Bioelectric Membrane Potentials: Implications in Cancer Pathogenesis and Therapeutic Strategies. J Membr Biol 2024:10.1007/s00232-024-00323-2. [PMID: 39183198 DOI: 10.1007/s00232-024-00323-2] [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: 05/30/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024]
Abstract
Electrophysiology typically deals with the electrical properties of excitable cells like neurons and muscles. However, all other cells (non-excitable) also possess bioelectric membrane potentials for intracellular and extracellular communications. These membrane potentials are generated by different ions present in fluids available in and outside the cell, playing a vital role in communication and coordination between the cell and its organelles. Bioelectric membrane potential variations disturb cellular ionic homeostasis and are characteristic of many diseases, including cancers. A rapidly increasing interest has emerged in sorting out the electrophysiology of cancer cells. Compared to healthy cells, the distinct electrical properties exhibited by cancer cells offer a unique way of understanding cancer development, migration, and progression. Decoding the altered bioelectric signals influenced by fluctuating electric fields benefits understanding cancer more closely. While cancer research has predominantly focussed on genetic and molecular traits, the delicate area of electrophysiological characteristics has increasingly gained prominence. This review explores the historical exploration of electrophysiology in the context of cancer cells, shedding light on how alterations in bioelectric membrane potentials, mediated by ion channels and gap junctions, contribute to the pathophysiology of cancer.
Collapse
Affiliation(s)
- Anju Shrivastava
- Department of Physiology, Chhattisgarh Institute of Medical Sciences, Bilaspur, India.
| | - Amit Kumar
- Department of Anatomy, Chhattisgarh Institute of Medical Sciences, Bilaspur, India
| | - Lalit Mohan Aggarwal
- Radiotherapy and Radiation Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Satyajit Pradhan
- Radiation Oncology, Mahamana Pandit Madhan Mohan Malaviya Cancer Centre, Varanasi, India
| | - Sunil Choudhary
- Radiotherapy and Radiation Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Ashish Ashish
- Department of Anatomy, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Keshav Kashyap
- Department of Physiology, Chhattisgarh Institute of Medical Sciences, Bilaspur, India
| | - Shivani Mishra
- Department of Anatomy, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
2
|
Min Q, Gao Y, Wang Y. Bioelectricity in dental medicine: a narrative review. Biomed Eng Online 2024; 23:3. [PMID: 38172866 PMCID: PMC10765628 DOI: 10.1186/s12938-023-01189-6] [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: 09/07/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Bioelectric signals, whether exogenous or endogenous, play crucial roles in the life processes of organisms. Recently, the significance of bioelectricity in the field of dentistry is steadily gaining greater attention. OBJECTIVE This narrative review aims to comprehensively outline the theory, physiological effects, and practical applications of bioelectricity in dental medicine and to offer insights into its potential future direction. It attempts to provide dental clinicians and researchers with an electrophysiological perspective to enhance their clinical practice or fundamental research endeavors. METHODS An online computer search for relevant literature was performed in PubMed, Web of Science and Cochrane Library, with the keywords "bioelectricity, endogenous electric signal, electric stimulation, dental medicine." RESULTS Eventually, 288 documents were included for review. The variance in ion concentration between the interior and exterior of the cell membrane, referred to as transmembrane potential, forms the fundamental basis of bioelectricity. Transmembrane potential has been established as an essential regulator of intercellular communication, mechanotransduction, migration, proliferation, and immune responses. Thus, exogenous electric stimulation can significantly alter cellular action by affecting transmembrane potential. In the field of dental medicine, electric stimulation has proven useful for assessing pulp condition, locating root apices, improving the properties of dental biomaterials, expediting orthodontic tooth movement, facilitating implant osteointegration, addressing maxillofacial malignancies, and managing neuromuscular dysfunction. Furthermore, the reprogramming of bioelectric signals holds promise as a means to guide organism development and intervene in disease processes. Besides, the development of high-throughput electrophysiological tools will be imperative for identifying ion channel targets and precisely modulating bioelectricity in the future. CONCLUSIONS Bioelectricity has found application in various concepts of dental medicine but large-scale, standardized, randomized controlled clinical trials are still necessary in the future. In addition, the precise, repeatable and predictable measurement and modulation methods of bioelectric signal patterns are essential research direction.
Collapse
Affiliation(s)
- Qingqing Min
- Department of Endodontics, Wuxi Stomatology Hospital, Wuxi, 214000, China
| | - Yajun Gao
- Department of Endodontics, Wuxi Stomatology Hospital, Wuxi, 214000, China
| | - Yao Wang
- Department of Implantology, Wuxi Stomatology Hospital, Wuxi, 214000, China.
| |
Collapse
|
3
|
Mishra L, Bhowmik S, Singh R, Patel P, Gupta GD, Kurmi BD. Quality by design-assisted development of D-α-tocopherol polyethylene glycol 1000 succinate-incorporated gefitinib-loaded cationic liposome(s). Ther Deliv 2023; 14:745-761. [PMID: 38018431 DOI: 10.4155/tde-2023-0075] [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] [Indexed: 11/30/2023] Open
Abstract
Aim: Gefitinib-loaded D-α-tocopherol polyethylene glycol 1000 succinate (TPGS)-coated cationic liposomes (GEF-TPGS-LIPO+) were developed and optimized by the quality by design (QbD) approach for its potential anticancer effect. Methods/materials: Box-Behnken design (BBD) a systematic design of experiments was added to screen and optimize the formulation variables. Results: GEF-TPGS-LIPO+ shows vesicle size (210 ± 4.82 nm), polydispersity index (0.271 ± 0.002), zeta potential (22.2 ± 0.84 mV) and entrapment efficiency (82.3 ± 1.95). MTT result shows the enhanced cytotoxicity and higher intracellular drug uptake with highest and lowest levels of the reactive oxygen species and NF-κB expressions on A549 lung cancer cells, determined by fluorescence-activated cell sorting flow cytometry. Conclusion: Potential anticancer effect on A549 cells might be found due to cationic liposomal interaction with cancer cells.
Collapse
Affiliation(s)
- Lopamudra Mishra
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, Punjab, 142001, India
| | - Shuvadip Bhowmik
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, Punjab, 142001, India
| | - Rajveer Singh
- Department of Pharmacognosy, ISF College Pharmacy, GT Road, Moga, Punjab, 142001, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College Pharmacy, GT Road, Moga, Punjab, 142001, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, Punjab, 142001, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, Punjab, 142001, India
| |
Collapse
|
4
|
Zhou X, Li G, Wu D, Liang H, Zhang W, Zeng L, Zhu Q, Lai P, Wen Z, Yang C, Pan Y. Recent advances of cellular stimulation with triboelectric nanogenerators. EXPLORATION (BEIJING, CHINA) 2023; 3:20220090. [PMID: 37933231 PMCID: PMC10624380 DOI: 10.1002/exp.20220090] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 12/06/2022] [Indexed: 11/08/2023]
Abstract
Triboelectric nanogenerators (TENGs) are new energy collection devices that have the characteristics of high efficiency, low cost, miniaturization capability, and convenient manufacture. TENGs mainly utilize the triboelectric effect to obtain mechanical energy from organisms or the environment, and this mechanical energy is then converted into and output as electrical energy. Bioelectricity is a phenomenon that widely exists in various cellular processes, including cell proliferation, senescence, apoptosis, as well as adjacent cells' communication and coordination. Therefore, based on these features, TENGs can be applied in organisms to collect energy and output electrical stimulation to act on cells, changing their activities and thereby playing a role in regulating cellular function and interfering with cellular fate, which can further develop into new methods of health care and disease intervention. In this review, we first introduce the working principle of TENGs and their working modes, and then summarize the current research status of cellular function regulation and fate determination stimulated by TENGs, and also analyze their application prospects for changing various processes of cell activity. Finally, we discuss the opportunities and challenges of TENGs in the fields of life science and biomedical engineering, and propose a variety of possibilities for their potential development direction.
Collapse
Affiliation(s)
- Xingyu Zhou
- Department of Orthopaedics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA MedicineMedical Research Center, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Gaocai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA MedicineMedical Research Center, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Di Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Huaizhen Liang
- Department of Orthopaedics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Weifeng Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Lingli Zeng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA MedicineMedical Research Center, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Qianqian Zhu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon‐Based Functional Materials and DevicesSoochow UniversitySuzhouChina
| | - Puxiang Lai
- Department of Biomedical EngineeringHong Kong Polytechnic UniversityHong KongChina
| | - Zhen Wen
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon‐Based Functional Materials and DevicesSoochow UniversitySuzhouChina
| | - Cao Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yue Pan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA MedicineMedical Research Center, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
5
|
Moreddu R, Boschi A, d’Amora M, Hubarevich A, Dipalo M, De Angelis F. Passive Recording of Bioelectrical Signals from Non-Excitable Cells by Fluorescent Mirroring. NANO LETTERS 2023; 23:3217-3223. [PMID: 37019439 PMCID: PMC10141418 DOI: 10.1021/acs.nanolett.2c05053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Bioelectrical variations trigger different cell responses, including migration, mitosis, and mutation. At the tissue level, these actions result in phenomena such as wound healing, proliferation, and pathogenesis. Monitoring these mechanisms dynamically is highly desirable in diagnostics and drug testing. However, existing technologies are invasive: either they require physical access to the intracellular compartments, or they imply direct contact with the cellular medium. Here, we present a novel approach for the passive recording of electrical signals from non-excitable cells adhering to 3D microelectrodes, based on optical mirroring. Preliminary results yielded a fluorescence intensity output increase of the 5,8% in the presence of a HEK-293 cell on the electrode compared to bare microelectrodes. At present, this technology may be employed to evaluate cell-substrate adhesion and monitor cell proliferation. Further refinements could allow extrapolating quantitative data on surface charges and resting potential to investigate the electrical phenomena involved in cell migration and cancer progression.
Collapse
Affiliation(s)
- Rosalia Moreddu
- Plasmon
Nanotechnologies Unit, Istituto Italiano
di Tecnologia, 16163 Genoa, Italy
| | - Alessio Boschi
- Plasmon
Nanotechnologies Unit, Istituto Italiano
di Tecnologia, 16163 Genoa, Italy
- Department
of Bioengineering, University of Genoa, 16126 Genoa, Italy
| | - Marta d’Amora
- Plasmon
Nanotechnologies Unit, Istituto Italiano
di Tecnologia, 16163 Genoa, Italy
- Department
of Biology, University of Pisa, 56127 Pisa, Italy
| | | | - Michele Dipalo
- Plasmon
Nanotechnologies Unit, Istituto Italiano
di Tecnologia, 16163 Genoa, Italy
- Email
| | - Francesco De Angelis
- Plasmon
Nanotechnologies Unit, Istituto Italiano
di Tecnologia, 16163 Genoa, Italy
- Email
| |
Collapse
|
6
|
Principle Superiority and Clinical Extensibility of 2D and 3D Charged Nanoprobe Detection Platform Based on Electrophysiological Characteristics of Circulating Tumor Cells. Cells 2023; 12:cells12020305. [PMID: 36672240 PMCID: PMC9856308 DOI: 10.3390/cells12020305] [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: 11/13/2022] [Revised: 12/21/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023] Open
Abstract
The electrical characteristic of cancer cells is neglected among tumor biomarkers. The development of nanoprobes with opposing charges for monitoring the unique electrophysiological characteristics of cancer cells. Micro-nano size adsorption binding necessitates consideration of the nanoprobe's specific surface area. On the basis of the electrophysiological characteristics of circulating tumor cells (CTCs), clinical application and performance assessment are determined. To demonstrate that cancer cells have a unique pattern of electrophysiological patterns compared to normal cells, fluorescent nanoprobes with opposing charges were developed and fabricated. Graphene oxide (GO) was used to transform three-dimensional (3D) nanoprobes into two-dimensional (2D) nanoprobes. Compare 2D and 3D electrophysiological magnetic nanoprobes (MNP) in clinical samples and evaluate the adaptability and development of CTCs detection based on cell electrophysiology. Positively charged nanoprobes rapidly bind to negatively charged cancer cells based on electrostatic interactions. Compared to MNPs(+) without GO, the GO/MNPs(+) nanoprobe is more efficient and uses less material to trap cancer cells. CTCs can be distinguished from normal cells that are fully unaffected by nanoprobes by microscopic cytomorphological inspection, enabling the tracking of the number and pathological abnormalities of CTCs in the same patient at various chemotherapy phases to determine the efficacy of treatment. The platform for recognizing CTCs on the basis of electrophysiological characteristics compensates for the absence of epithelial biomarker capture and size difference capture in clinical performance. Under the influence of electrostatic attraction, the binding surface area continues to influence the targeting of cancer cells by nanoprobes. The specific recognition and detection of nanoprobes based on cell electrophysiological patterns has enormous potential in the clinical diagnosis and therapeutic monitoring of cancer.
Collapse
|
7
|
Bory Prevez H, Soutelo Jimenez AA, Roca Oria EJ, Heredia Kindelán JA, Morales González M, Villar Goris NA, Hernández Mesa N, Sierra González VG, Infantes Frometa Y, Montijano JI, Cabrales LEB. Simulations of surface charge density changes during the untreated solid tumour growth. ROYAL SOCIETY OPEN SCIENCE 2022; 9:220552. [PMID: 36465673 PMCID: PMC9709566 DOI: 10.1098/rsos.220552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 11/10/2022] [Indexed: 06/17/2023]
Abstract
Understanding untreated tumour growth kinetics and its intrinsic behaviour is interesting and intriguing. The aim of this study is to propose an approximate analytical expression that allows us to simulate changes in surface charge density at the cancer-surrounding healthy tissue interface during the untreated solid tumour growth. For this, the Gompertz and Poisson equations are used. Simulations reveal that the unperturbed solid tumour growth is closely related to changes in the surface charge density over time between the tumour and the surrounding healthy tissue. Furthermore, the unperturbed solid tumour growth is governed by temporal changes in this surface charge density. It is concluded that results corroborate the correspondence between the electrical and physiological parameters in the untreated cancer, which may have an essential role in its growth, progression, metastasis and protection against immune system attack and anti-cancer therapies. In addition, the knowledge of surface charge density changes at the cancer-surrounding healthy tissue interface may be relevant when redesigning the molecules in chemotherapy and immunotherapy taking into account their polarities. This can also be true in the design of completely novel therapies.
Collapse
Affiliation(s)
- Henry Bory Prevez
- Departamento de Control Automático, Facultad de Ingeniería Eléctrica, Universidad de Oriente, Santiago de Cuba, Cuba
| | | | - Eduardo José Roca Oria
- Departamento de Física, Facultad de Ciencias Naturales y Exactas, Universidad de Oriente, Santiago de Cuba, Cuba
| | | | - Maraelys Morales González
- Departamento de Farmacia, Facultad de Ciencias Naturales y Exactas, Universidad de Oriente, Santiago de Cuba, Cuba
| | - Narciso Antonio Villar Goris
- Departamento de Ciencia e Innovación, Centro Nacional de Electromagnetismo Aplicado, Universidad de Oriente, Santiago de Cuba, Cuba
- Universidad Autónoma de Santo Domingo, Santo Domingo, República Dominicana
| | | | | | | | - Juan Ignacio Montijano
- Departamento de Matemática Aplicada, Instituto Universitario de Matemática y Aplicaciones, Universidad de Zaragoza, Zaragoza, España
| | - Luis Enrique Bergues Cabrales
- Departamento de Ciencia e Innovación, Centro Nacional de Electromagnetismo Aplicado, Universidad de Oriente, Santiago de Cuba, Cuba
- Departamento de Matemática Aplicada, Instituto Universitario de Matemática y Aplicaciones, Universidad de Zaragoza, Zaragoza, España
| |
Collapse
|
8
|
Deng Z, Wu S, Wang Y, Shi D. Circulating tumor cell isolation for cancer diagnosis and prognosis. EBioMedicine 2022; 83:104237. [PMID: 36041264 PMCID: PMC9440384 DOI: 10.1016/j.ebiom.2022.104237] [Citation(s) in RCA: 97] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 11/03/2022] Open
Abstract
Circulating tumor cells (CTCs) are tumor cells that shed from the primary tumor and intravasate into the peripheral blood circulation system responsible for metastasis. Sensitive detection of CTCs from clinical samples can serve as an effective tool in cancer diagnosis and prognosis through liquid biopsy. Current CTC detection technologies mainly reply on the biomarker-mediated platforms including magnetic beads, microfluidic chips or size-sensitive microfiltration which can compromise detection sensitivity due to tumor heterogeneity. A more sensitive, biomarker independent CTCs isolation technique has been recently developed with the surface-charged superparamagnetic nanoprobe capable of different EMT subpopulation CTC capture from 1 mL clinical blood. In this review, this new strategy is compared with the conventional techniques on biomarker specificity, impact of protein corona, effect of glycolysis on cell surface charge, and accurate CTC identification. Correlations between CTC enumeration and molecular profiling in clinical blood and cancer prognosis are provided for clinical cancer management.
Collapse
Affiliation(s)
- Zicheng Deng
- The Materials Science and Engineering Program, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH 45221, USA; Center for Lung Regenerative Medicine, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Shengming Wu
- The Institute for Translational Nanomedicine Shanghai East Hospital, The Institute for Biomedical Engineering and Nano Science, School of Medicine Tongji University, Shanghai 200092, PR China
| | - Yilong Wang
- The Institute for Translational Nanomedicine Shanghai East Hospital, The Institute for Biomedical Engineering and Nano Science, School of Medicine Tongji University, Shanghai 200092, PR China.
| | - Donglu Shi
- The Materials Science and Engineering Program, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH 45221, USA.
| |
Collapse
|
9
|
Ju S, Chen C, Zhang J, Xu L, Zhang X, Li Z, Chen Y, Zhou J, Ji F, Wang L. Detection of circulating tumor cells: opportunities and challenges. Biomark Res 2022; 10:58. [PMID: 35962400 PMCID: PMC9375360 DOI: 10.1186/s40364-022-00403-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/26/2022] [Indexed: 11/25/2022] Open
Abstract
Circulating tumor cells (CTCs) are cells that shed from a primary tumor and travel through the bloodstream. Studying the functional and molecular characteristics of CTCs may provide in-depth knowledge regarding highly lethal tumor diseases. Researchers are working to design devices and develop analytical methods that can capture and detect CTCs in whole blood from cancer patients with improved sensitivity and specificity. Techniques using whole blood samples utilize physical prosperity, immunoaffinity or a combination of the above methods and positive and negative enrichment during separation. Further analysis of CTCs is helpful in cancer monitoring, efficacy evaluation and designing of targeted cancer treatment methods. Although many advances have been achieved in the detection and molecular characterization of CTCs, several challenges still exist that limit the current use of this burgeoning diagnostic approach. In this review, a brief summary of the biological characterization of CTCs is presented. We focus on the current existing CTC detection methods and the potential clinical implications and challenges of CTCs. We also put forward our own views regarding the future development direction of CTCs.
Collapse
Affiliation(s)
- Siwei Ju
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Cong Chen
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Jiahang Zhang
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Lin Xu
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Xun Zhang
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Zhaoqing Li
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Yongxia Chen
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Jichun Zhou
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China
| | - Feiyang Ji
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China.
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China.
| | - Linbo Wang
- Department of Surgical Oncology, The Sir Run Run Shaw Hospital Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China.
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang, Hangzhou, China.
| |
Collapse
|
10
|
Co-Delivery of 5-Fluorouracil and Paclitaxel in Mitochondria-Targeted KLA-Modified Liposomes to Improve Triple-Negative Breast Cancer Treatment. Pharmaceuticals (Basel) 2022; 15:ph15070881. [PMID: 35890181 PMCID: PMC9318860 DOI: 10.3390/ph15070881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 11/19/2022] Open
Abstract
In this research, KLA-modified liposomes co-loaded with 5-fluorouracil and paclitaxel (KLA-5-FU/PTX Lps) were developed, and their antitumor activity against triple-negative breast cancer (TNBC) was evaluated. KLA-5-FU/PTX Lps were prepared using the thin-film dispersion method, and their in vitro anticancer efficacy was assessed in human breast cancer cells (MDA-MB-231). An MDA-MB-231 tumor-bearing mouse model was also established to evaluate their antitumor efficacy in vivo. KLA-5-FU/PTX Lps showed enhanced cytotoxicity against MDA-MB-231 cells, improved drug delivery to mitochondria, and induced mitochondria-mediated apoptosis. The modified liposomes also showed favorable antitumor activity in vivo due to their strong ability to target tumors and mitochondria. The liposomes showed no obvious systemic toxicity. Our results suggest that KLA-5-FU/PTX Lps are a promising system with which to target the delivery of antitumor drugs to mitochondria as a treatment for TNBC.
Collapse
|
11
|
Dual Targeting with Cell Surface Electrical Charge and Folic Acid via Superparamagnetic Fe 3O 4@Cu 2-xS for Photothermal Cancer Cell Killing. Cancers (Basel) 2021; 13:cancers13215275. [PMID: 34771438 PMCID: PMC8582571 DOI: 10.3390/cancers13215275] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 09/28/2021] [Accepted: 10/19/2021] [Indexed: 12/27/2022] Open
Abstract
Simple Summary There are two critical issues in cancer hyperthermia: (1) photothermal effect and (2) cancer cell targeting efficiency. While the former can be addressed by rendering the nano carriers with significant IR absorptions, the latter is dealt with using a novel dual-targeting strategy. In this study, the Fe3O4 nanoparticle was coated with a shell of Cu2–xS; the resulting Fe3O4@Cu2–xS exhibited strong IR absorption for enhanced photothermal cancer cell killing. The Fe3O4@Cu2–xS nanoparticles are surface functionalized with amphiphilic polyethylenimine (LA-PEI) and Folic acid-TPGS (FA-TPGS) for two purposes: (1) the PEI surface coating renders the particles positively charged, enabling them to effectively bind with negatively-charged cancer cells for more intimate nano/bio contact resulting in much stronger cancer cell ablation; (2) the folic acid modification further increases the targeting efficiency via the folic receptors on the cancer cell surface. Dual-targeting with the surface electrical charge and the tumor-specific folic acid synergistically facilitates both passive and active targeting for significantly improved photothermal killing. Abstract A major challenge in cancer therapy is to achieve high cell targeting specificity for the highest therapeutic efficacy. Two major approaches have been shown to be quite effective, namely, (1) bio-marker mediated cell targeting, and (2) electrical charge driven cell binding. The former utilizes the tumor-specific moieties on nano carrier surfaces for active targeting, while the latter relies on nanoparticles binding onto the cancer cell surfaces due to differences in electrical charge. Cancer cells are known for their hallmark metabolic pattern: high rates of glycolysis that lead to negatively charged cell surfaces. In this study, the nanoparticles of Fe3O4@Cu2–xS were rendered positively charged by conjugating their surfaces with different functional groups for strong electrostatic binding onto the negatively-charged cancer cells. In addition to the positively charged surfaces, the Fe3O4@Cu2–xS nanoparticles were also modified with folic acid (FA) for biomarker-based cell targeting. The dual-targeting approach synergistically utilizes the effectiveness of both charge- and biomarker-based cell binding for enhanced cell targeting. Further, these superparamagnetic Fe3O4@Cu2–xS nanoparticles exhibit much stronger IR absorptions compared to Fe3O4, therefore much more effective in photothermal therapy.
Collapse
|
12
|
Phillips JA, Hutchings C, Djamgoz MBA. Clinical Potential of Nerve Input to Tumors: A Bioelectricity Perspective. Bioelectricity 2021; 3:14-26. [PMID: 34476375 DOI: 10.1089/bioe.2020.0051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
We support the notion that the neural connections of the tumor microenvironment (TME) and the associated 'bioelectricity' play significant role in the pathophysiology of cancer. In several cancers, the nerve input promotes the cancer process. While straightforward surgical denervation of tumors, therefore, could improve prognosis, resulting side effects of such a procedure would be unpredictable and irreversible. On the other hand, tumor innervation can be manipulated effectively for therapeutic purposes by alternative novel approaches broadly termed "electroceuticals." In this perspective, we evaluate the clinical potential of targeting the TME first through manipulation of the nerve input itself and second by application of electric fields directly to the tumor. The former encompasses several different biophysical and biochemical approaches. These include implantable devices, nanoparticles, and electroactive polymers, as well as optogenetics and chemogenetics. As regard bioelectrical manipulation of the tumor itself, the "tumor-treating field" technique, applied to gliomas commonly in combination with chemotherapy, is evaluated. Also, as electroceuticals, drugs acting on ion channels and neurotransmitter receptors are highlighted for completeness. It is concluded, first, that electroceuticals comprise a broad range of biomedical tools. Second, such electroceuticals present significant clinical potential for exploiting the neural component of the TME as a strategy against cancer. Finally, the inherent bioelectric characteristics of tumors themselves are also amenable to complementary approaches. Collectively, these represent an evolving, dynamic field and further progress and applications can be expected to follow both conceptually and technically.
Collapse
Affiliation(s)
- Jade A Phillips
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Charlotte Hutchings
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Mustafa B A Djamgoz
- Department of Life Sciences, Imperial College London, London, United Kingdom.,Biotechnology Research Center, Cyprus International University, Nicosia, North Cyprus
| |
Collapse
|
13
|
Li Z, Liu X, Zhang W, Zhuang X. Electrostatic reaction for the detection of circulating tumor cells as a potential diagnostic biomarker for metastasis in solid tumor. Nanotheranostics 2020; 4:233-241. [PMID: 32923313 PMCID: PMC7484632 DOI: 10.7150/ntno.46928] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/28/2020] [Indexed: 12/24/2022] Open
Abstract
The detection of circulating tumor cells (CTCs) from blood samples is important to predict metastatic spread of cancer cells. However, effective quantification and identification of CTCs in solid tumors remain a challenge. Aerobic glycolysis is a hallmark of cancer cells, which makes cancer cells have more negative membrane potentials than that of normal cells. Herein, we reported a CTC isolation method with 80.7% capture efficiency based on electrostatic reaction, which was accomplished within 30 min in mimic clinical samples. Following in vitro verification using Lewis lung carcinoma (LLC1) (EpCAM-positive) and B16F10 (EpCAM-negative) melanoma cells, syngeneic tumor models were used to evaluate specificity and sensitivity of the surface charged nanoparticles. After subcutaneous implantation, blood was drawn from mice every three days, and CTCs were successfully detected in all implanted subjects. From 100 µl blood samples, the minimum amount found in blood was 9-34 CTCs on 3 day and the maximum was 94-107 CTCs on 15 day. Besides, the isolated CTCs cells remained viable and verified by re-implantation. This study confirms that our multifunctional nanoparticles are highly efficient in detecting CTCs in tumor metastasis and has huge potential in translational medicine.
Collapse
Affiliation(s)
- Zhiming Li
- Institue of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.,Department of Urology, the First Affiliated Hospital of Xiamen University, Xiamen 361003, Fujian, China
| | - Xingping Liu
- Institue of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Weidong Zhang
- Clinical Trial Management Platform, Jinhua Municipal Central Hospital, Jinhua 321000, Zhejiang, China
| | - Xuan Zhuang
- Department of Urology, the First Affiliated Hospital of Xiamen University, Xiamen 361003, Fujian, China.,Department of Clinical Medicine, Fujian Medical University, Fuzhou 350005, Fujian Province, China
| |
Collapse
|
14
|
Li C, Li Y, Yao T, Zhou L, Xiao C, Wang Z, Zhai J, Xing J, Chen J, Tan G, Zhou Y, Qi S, Yu P, Ning C. Wireless Electrochemotherapy by Selenium-Doped Piezoelectric Biomaterials to Enhance Cancer Cell Apoptosis. ACS APPLIED MATERIALS & INTERFACES 2020; 12:34505-34513. [PMID: 32508084 DOI: 10.1021/acsami.0c04666] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Cancer residues around the surgical site remain a significant cause of treatment failure with cancer recurrence. To prevent cancer recurrence and simultaneously repair surgery-caused defects, it is urgent to develop implantable biomaterials with anticancer ability and good biological activity. In this work, a functionalized implant is successfully fabricated by doping the effective anticancer element selenium (Se) into the potassium-sodium niobate piezoceramic, which realizes the wireless combination of electrotherapy and chemotherapy. Herein, we demonstrate that the Se-doped piezoelectric implant can cause mitochondrial damage by increasing intracellular reactive oxygen species levels and then trigger the caspase-3 pathway to significantly promote apoptosis of osteosarcoma cells in vitro. Meanwhile, its good biocompatibility has been verified. These results are of great importance for future deployment of wireless electro- and chemostimulation to modulate biological process around the defective tissue.
Collapse
Affiliation(s)
- Changhao Li
- School of Material Science and Engineering, South China University of Technology, Guangzhou 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P. R. China
| | - Yangfan Li
- School of Material Science and Engineering, South China University of Technology, Guangzhou 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P. R. China
| | - Tiantian Yao
- School of Material Science and Engineering, South China University of Technology, Guangzhou 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P. R. China
| | - Lei Zhou
- School of Material Science and Engineering, South China University of Technology, Guangzhou 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P. R. China
| | - Cairong Xiao
- School of Material Science and Engineering, South China University of Technology, Guangzhou 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P. R. China
| | - Zhengao Wang
- School of Material Science and Engineering, South China University of Technology, Guangzhou 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P. R. China
| | - Jinxia Zhai
- School of Material Science and Engineering, South China University of Technology, Guangzhou 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P. R. China
| | - Jun Xing
- School of Material Science and Engineering, South China University of Technology, Guangzhou 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P. R. China
| | - Junqi Chen
- School of Material Science and Engineering, South China University of Technology, Guangzhou 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P. R. China
| | - Guoxin Tan
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, P. R. China
| | - Yahong Zhou
- CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Science, Beijing 100190, China
| | - Suijian Qi
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Peng Yu
- School of Material Science and Engineering, South China University of Technology, Guangzhou 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P. R. China
| | - Chengyun Ning
- School of Material Science and Engineering, South China University of Technology, Guangzhou 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P. R. China
| |
Collapse
|
15
|
Ni L, Shaik R, Xu R, Zhang G, Zhe J. A Microfluidic Sensor for Continuous, in Situ Surface Charge Measurement of Single Cells. ACS Sens 2020; 5:527-534. [PMID: 31939290 DOI: 10.1021/acssensors.9b02411] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Cell surface charge has been recognized as an important cellular property. We developed a microfluidic sensor based on resistive pulse sensing to assess surface charge and sizes of single cells suspended in a continuous flow. The device consists of two consecutive resistive pulse sensors (RPSs) with identical dimensions. Opposite electric fields were applied on the two RPSs. A charged cell in the RPSs was accelerated or decelerated by the electric fields and thus exhibited different transit times passing through the two RPSs. The cell surface charge is measured with zeta potential that can be quantified with the transit time difference. The transit time of each cell can be accurately detected with the width of pulses generated by the RPS, while the cell size can be calculated with the pulse magnitude at the same time. This device has the ability to detect surface charges and sizes of individual cells with high tolerance in cell types and testing solutions compared with traditional electrophoretic light scattering methods. Three different types of cells including HeLa cancer cells, human dermal fibroblast cells, and human umbilical vein endothelial cells (HUVECs) were tested with the sensor. Results showed a significant difference of zeta potentials between HeLa cells and fibroblasts or HUVECs. In addition, when HeLa cells were treated with various concentrations of glutamine, the effects on cancer cell surface charge were detected. Our results demonstrated the great potential of using our sensor for cell type sorting, cancer cell detection, and cell status analysis.
Collapse
Affiliation(s)
- Liwei Ni
- Department of Mechanical Engineering, University of Akron, Akron, Ohio 44325, United States
| | - Rubia Shaik
- Department of Biomedical Engineering, University of Akron, Akron, Ohio 44325, United States
| | - Ruiting Xu
- Department of Mechanical Engineering, University of Akron, Akron, Ohio 44325, United States
| | - Ge Zhang
- Department of Biomedical Engineering, University of Akron, Akron, Ohio 44325, United States
| | - Jiang Zhe
- Department of Mechanical Engineering, University of Akron, Akron, Ohio 44325, United States
| |
Collapse
|
16
|
Wu S, Gu L, Qin J, Zhang L, Sun F, Liu Z, Wang Y, Shi D. Rapid Label-Free Isolation of Circulating Tumor Cells from Patients' Peripheral Blood Using Electrically Charged Fe 3O 4 Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2020; 12:4193-4203. [PMID: 31935069 DOI: 10.1021/acsami.9b16385] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Isolation of circulating tumor cells (CTCs) in peripheral blood from cancer patients bears critical importance for evaluation of therapeutic efficacy. The current CTC isolation strategies are majorly relying on either protein biomarkers or dimensional features of CTCs. In this study, we present a new methodology for CTC detection and isolation based on the surface charge of cancer cells, a bioelectrical manifestation of the "Warburg effect." Negative surface charge is a direct consequence of glycolysis of cancer cells, which can be utilized as an effective biophysical marker for CTC detection and isolation. Upon cancer cells-nanoparticle interaction via optimum incubation, serum protein-coated electrically charged nanoparticles can trap different cancer cells independent of their epithelial protein expression. In fetal bovine serum , the poly(ethyleneimine)-functionalized Fe3O4 nanoparticles, surface-decorated with protein corona, are able to efficiently capture CTCs from blood samples of colorectal cancer patients. 2-8 CTCs has been isolated from 1 mL of blood and identified by immunostaining fluorescence in situ hybridization and immunofluorescence staining in all 25 colorectal cancer patients at varied stages, while only 0-1 CTC was detected from blood samples of 10 healthy donors. Diverse CTC subpopulations of heteroploids and biomarker expression can also be detected in this strategy. The label-free, charge-based CTC method shows promise in cancer diagnosis and prognosis paving a new path for liquid biopsy.
Collapse
Affiliation(s)
- Shengming Wu
- The Institute for Translational Nanomedicine, Shanghai East Hospital, the Institute for Biomedical Engineering & Nano Science , Tongji University School of Medicine , Shanghai 200092 , P. R. China
| | - Lei Gu
- Department of General Surgery , Shanghai Tenth People's Hospital of Tongji University School of Medicine , Shanghai 200072 , P. R. China
| | - Jingwen Qin
- The Institute for Translational Nanomedicine, Shanghai East Hospital, the Institute for Biomedical Engineering & Nano Science , Tongji University School of Medicine , Shanghai 200092 , P. R. China
| | - Lei Zhang
- Department of Clinical Laboratory Medicine , Shanghai Tenth People's Hospital of Tongji University , Shanghai 200072 , P. R. China
| | - Fenyong Sun
- Department of Clinical Laboratory Medicine , Shanghai Tenth People's Hospital of Tongji University , Shanghai 200072 , P. R. China
| | - Zhongchen Liu
- Department of General Surgery , Shanghai Tenth People's Hospital of Tongji University School of Medicine , Shanghai 200072 , P. R. China
| | - Yilong Wang
- The Institute for Translational Nanomedicine, Shanghai East Hospital, the Institute for Biomedical Engineering & Nano Science , Tongji University School of Medicine , Shanghai 200092 , P. R. China
| | - Donglu Shi
- The Materials Science and Engineering Program, Dept. of Mechanical and Materials Engineering, College of Engineering and Applied Science , University of Cincinnati , Cincinnati , Ohio 45221 , United States
| |
Collapse
|