1
|
Elnagar N, Elgiddawy N, El Rouby WMA, Farghali AA, Korri-Youssoufi H. Impedimetric Detection of Cancer Markers Based on Nanofiber Copolymers. BIOSENSORS 2024; 14:77. [PMID: 38391996 PMCID: PMC10887276 DOI: 10.3390/bios14020077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/23/2024] [Accepted: 01/29/2024] [Indexed: 02/24/2024]
Abstract
The sensitive determination of folate receptors (FRs) in the early stages of cancer is of great significance for controlling the progression of cancerous cells. Many folic acid (FA)-based electrochemical biosensors have been utilized to detect FRs with promising performances, but most were complicated, non-reproducible, non-biocompatible, and time and cost consuming. Here, we developed an environmentally friendly and sensitive biosensor for FR detection. We proposed an electrochemical impedimetric biosensor formed by nanofibers (NFs) of bio-copolymers prepared by electrospinning. The biosensor combines the advantages of bio-friendly polymers, such as sodium alginate (SA) and polyethylene oxide (PEO) as an antifouling polymer, with FA as a biorecognition element. The NF nanocomposites were characterized using various techniques, including SEM, FTIR, zeta potential (ZP), cyclic voltammetry (CV), and electrochemical impedance spectroscopy (EIS). We evaluated the performance of the NF biosensor using EIS and demonstrated FR detection in plasma with a limit of detection of 3 pM. Furthermore, the biosensor showed high selectivity, reliability, and good stability when stored for two months. This biosensor was constructed from 'green credentials' holding polymers that are highly needed in the new paradigm shift in the medical industry.
Collapse
Affiliation(s)
- Noha Elnagar
- Materials Science and Nanotechnology Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 62 511, Egypt; (N.E.); (W.M.A.E.R.); (A.A.F.)
- Université Paris-Saclay, Centre National de la Recherche Scientifique (CNRS), Institut de Chimie Moléculaire et des Matériaux d’Orsay (ICMMO), ECBB, 17 Avenue des Sciences, Site Henri Moisson, 91400 Orsay, France
| | - Nada Elgiddawy
- Department of Biotechnology and Life Sciences, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 62 511, Egypt;
| | - Waleed M. A. El Rouby
- Materials Science and Nanotechnology Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 62 511, Egypt; (N.E.); (W.M.A.E.R.); (A.A.F.)
| | - Ahmed A. Farghali
- Materials Science and Nanotechnology Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 62 511, Egypt; (N.E.); (W.M.A.E.R.); (A.A.F.)
| | - Hafsa Korri-Youssoufi
- Université Paris-Saclay, Centre National de la Recherche Scientifique (CNRS), Institut de Chimie Moléculaire et des Matériaux d’Orsay (ICMMO), ECBB, 17 Avenue des Sciences, Site Henri Moisson, 91400 Orsay, France
| |
Collapse
|
2
|
Di Tucci C, Muzii L. Chronic Pelvic Pain, Vulvar Pain Disorders, and Proteomics Profiles: New Discoveries, New Hopes. Biomedicines 2023; 12:1. [PMID: 38275362 PMCID: PMC10813718 DOI: 10.3390/biomedicines12010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 01/27/2024] Open
Abstract
Chronic pelvic pain (CPP) is generally defined as non-cyclic pain perceived in the pelvic area that has persisted from three to six months or longer and is unrelated to pregnancy. The etiology of CPP is complex, multifactorial, with heterogeneous presentation, and includes several diseases such as endometriosis, adenomyosis, and interstitial cystitis/bladder pain syndrome. It may also be associated with sexual dysfunction, musculoskeletal disorders, and comorbid psychiatric symptoms. Vulvar pain disorders (VPDs) are typically categorized separately from chronic pelvic pain; among all VPDs, vulvodynia is a chronic vulvar pain of unknown etiology, lasting at least 3 months and that might be associated with other potentially linked factors. Proteomics represents a useful approach to study the proteome profiles of clinical samples. In this review, we have considered a selection of articles that have analyzed the protein abundance and novel protein species from various biological samples, including eutopic/ectopic endometrium, urine, serum, follicular, peritoneal fluid, and cervical mucus, potentially involved in the pathogenesis and progression of CPP and VPDs. These findings could represent valuable targets for paving the way for the differential diagnosis and therapeutic management of CPP and VDPs, thereby optimizing both the prevention and treatment of these conditions.
Collapse
Affiliation(s)
- Chiara Di Tucci
- Department of Obstetrics and Gynecology, “Sapienza” University, 00185 Rome, Italy;
| | | |
Collapse
|
3
|
Rizzo C, Cancemi P, Buttacavoli M, Di Cara G, D'Amico C, Billeci F, Marullo S, D'Anna F. Insights about the ability of folate based supramolecular gels to act as targeted therapeutic agents. J Mater Chem B 2023; 11:7721-7738. [PMID: 37466082 DOI: 10.1039/d3tb01389h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
With the aim to obtain targeted chemotherapeutic agents, imidazolium and ammonium-based folate salts were synthesized. Their photophysical behavior was investigated both in buffer and buffer/DMSO solution as well as in solid phase, performing UV-vis and fluorescence investigations. Properties of the aggregates were also analyzed by dynamic light scattering. Gelation ability of the salts was analyzed in biocompatible solvents, and gel phases obtained were characterized by determining critical gelation concentrations and gel-solution transition temperatures. Insights about gelator interactions in the tridimensional network were also gained performing ATR-FTIR investigation. Properties of soft materials were further analyzed performing rheology measurements, scanning electron microscopy, fluorescence and resonance light scattering investigations. Antiproliferative activity of organic salts was tested towards two breast cancer cell lines, expressing different levels of folate receptor, namely MDA-MB-231 and MCF-7, and a normal epithelial cell line, like h-TER T-RPE-1, by using MTT assay. Dichlodihydrofluorescein acetate test was performed to verify the role of oxidative stress in cell death. Finally, antiproliferative activity was also evaluated in gel phase, to verify if salts were able to retain biological activity also after the entrapment in the gelatinous network. Results collected evidence that folate based organic salts were able to behave as targeted chemotherapeutic agents both in solution and gel phase, showing uptake mechanism and selectivity indexes that depend on both cancer cell line nature and salt structure.
Collapse
Affiliation(s)
- Carla Rizzo
- Università degli Studi di Palermo, Dipartimento STEBICEF, Sezione di Chimica, Viale delle Scienze Ed. 17, 90128 Palermo, Italy.
| | - Patrizia Cancemi
- Università degli Studi di Palermo, Dipartimento STEBICEF, Sezione di Biologia Cellulare, Viale delle Scienze Ed. 16, 90128 Palermo, Italy
| | - Miriam Buttacavoli
- Università degli Studi di Palermo, Dipartimento STEBICEF, Sezione di Biologia Cellulare, Viale delle Scienze Ed. 16, 90128 Palermo, Italy
| | - Gianluca Di Cara
- Università degli Studi di Palermo, Dipartimento STEBICEF, Sezione di Biologia Cellulare, Viale delle Scienze Ed. 16, 90128 Palermo, Italy
| | - Cesare D'Amico
- Università degli Studi di Palermo, Dipartimento STEBICEF, Sezione di Biologia Cellulare, Viale delle Scienze Ed. 16, 90128 Palermo, Italy
| | - Floriana Billeci
- Università degli Studi di Palermo, Dipartimento STEBICEF, Sezione di Chimica, Viale delle Scienze Ed. 17, 90128 Palermo, Italy.
| | - Salvatore Marullo
- Università degli Studi di Palermo, Dipartimento STEBICEF, Sezione di Chimica, Viale delle Scienze Ed. 17, 90128 Palermo, Italy.
| | - Francesca D'Anna
- Università degli Studi di Palermo, Dipartimento STEBICEF, Sezione di Chimica, Viale delle Scienze Ed. 17, 90128 Palermo, Italy.
| |
Collapse
|
4
|
Neagu AN, Whitham D, Seymour L, Haaker N, Pelkey I, Darie CC. Proteomics-Based Identification of Dysregulated Proteins and Biomarker Discovery in Invasive Ductal Carcinoma, the Most Common Breast Cancer Subtype. Proteomes 2023; 11:13. [PMID: 37092454 PMCID: PMC10123686 DOI: 10.3390/proteomes11020013] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 04/05/2023] Open
Abstract
Invasive ductal carcinoma (IDC) is the most common histological subtype of malignant breast cancer (BC), and accounts for 70-80% of all invasive BCs. IDC demonstrates great heterogeneity in clinical and histopathological characteristics, prognoses, treatment strategies, gene expressions, and proteomic profiles. Significant proteomic determinants of the progression from intraductal pre-invasive malignant lesions of the breast, which characterize a ductal carcinoma in situ (DCIS), to IDC, are still poorly identified, validated, and clinically applied. In the era of "6P" medicine, it remains a great challenge to determine which patients should be over-treated versus which need to be actively monitored without aggressive treatment. The major difficulties for designating DCIS to IDC progression may be solved by understanding the integrated genomic, transcriptomic, and proteomic bases of invasion. In this review, we showed that multiple proteomics-based techniques, such as LC-MS/MS, MALDI-ToF MS, SELDI-ToF-MS, MALDI-ToF/ToF MS, MALDI-MSI or MasSpec Pen, applied to in-tissue, off-tissue, BC cell lines and liquid biopsies, improve the diagnosis of IDC, as well as its prognosis and treatment monitoring. Classic proteomics strategies that allow the identification of dysregulated protein expressions, biological processes, and interrelated pathway analyses based on aberrant protein-protein interaction (PPI) networks have been improved to perform non-invasive/minimally invasive biomarker detection of early-stage IDC. Thus, in modern surgical oncology, highly sensitive, rapid, and accurate MS-based detection has been coupled with "proteome point sampling" methods that allow for proteomic profiling by in vivo "proteome point characterization", or by minimal tissue removal, for ex vivo accurate differentiation and delimitation of IDC. For the detection of low-molecular-weight proteins and protein fragments in bodily fluids, LC-MS/MS and MALDI-MS techniques may be coupled to enrich and capture methods which allow for the identification of early-stage IDC protein biomarkers that were previously invisible for MS-based techniques. Moreover, the detection and characterization of protein isoforms, including posttranslational modifications of proteins (PTMs), is also essential to emphasize specific molecular mechanisms, and to assure the early-stage detection of IDC of the breast.
Collapse
Affiliation(s)
- Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iasi, Carol I bvd. No. 20A, 700505 Iasi, Romania
| | - Danielle Whitham
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | - Logan Seymour
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | - Norman Haaker
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | - Isabella Pelkey
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| | - Costel C. Darie
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA
| |
Collapse
|
5
|
Young O, Ngo N, Lin L, Stanbery L, Creeden JF, Hamouda D, Nemunaitis J. Folate Receptor as a Biomarker and Therapeutic Target in Solid Tumors. Curr Probl Cancer 2023; 47:100917. [PMID: 36508886 DOI: 10.1016/j.currproblcancer.2022.100917] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 10/21/2022] [Accepted: 10/27/2022] [Indexed: 11/18/2022]
Abstract
Folate is a B vitamin necessary for basic biological functions, including rapid cell turnover occurring in cancer cell proliferation. Though the role of folate as a causative versus protective agent in carcinogenesis is debated, several studies have indicated that the folate receptor (FR), notably subtype folate receptor alpha (FRα), could be a viable biomarker for diagnosis, progression, and prognosis. Several cancers, including gastrointestinal, gynecological, breast, lung, and squamous cell head and neck cancers overexpress FR and are currently under investigation to correlate receptor status to disease state. Traditional chemotherapies have included antifolate medications, such as methotrexate and pemetrexed, which generate anticancer activity during the synthesis phase of the cell cycle. Increasingly, the repertoire of pharmacotherapies is expanding to include FR as a target, with a heterogenous pool of directed therapies. Here we discuss the FR, expression and effect in cancer biology, and relevant pharmacologic inhibitors.
Collapse
Affiliation(s)
- Olivia Young
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Nealie Ngo
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Leslie Lin
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | | | - Justin Fortune Creeden
- Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Danae Hamouda
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | | |
Collapse
|
6
|
Carborane-Containing Folic Acid bis-Amides: Synthesis and In Vitro Evaluation of Novel Promising Agents for Boron Delivery to Tumour Cells. Int J Mol Sci 2022; 23:ijms232213726. [PMID: 36430206 PMCID: PMC9692863 DOI: 10.3390/ijms232213726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
The design of highly selective low-toxic, low-molecular weight agents for boron delivery to tumour cells is of decisive importance for the development of boron neutron capture therapy (BNCT), a modern efficient combined method for cancer treatment. In this work, we developed a simple method for the preparation of new closo- and nido-carborane-containing folic acid bis-amides containing 18-20 boron atoms per molecule. Folic acid derivatives containing nido-carborane residues were characterised by high water solubility, low cytotoxicity, and demonstrated a good ability to deliver boron to tumour cells in in vitro experiments (up to 7.0 µg B/106 cells in the case of U87 MG human glioblastoma cells). The results obtained demonstrate the high potential of folic acid-nido-carborane conjugates as boron delivery agents to tumour cells for application in BNCT.
Collapse
|
7
|
Sadeghi S, Homayouni Tabrizi M, Farhadi A. Folic acid-Chitosan Coated Stylosin Nanostructured Lipid Carriers: Fabrication, In Vitro-In Vivo Assessment in Breast Malignant Cells. JOURNAL OF BIOMATERIALS SCIENCE, POLYMER EDITION 2022; 34:791-809. [PMID: 36345914 DOI: 10.1080/09205063.2022.2145868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Synthesis of targeted nanostructure lipid carriers for stylosin (STY-CFN-NPs) delivery to MCF-7 cells. STY-CFN-NPs were formulated via the homogenization and ultra-sonication technique. After evaluating the amount of drug encapsulation and FA binding, the toxicity effect of the STY and STY-CFN-NPs on MCF-7 cells was measured by the MTT method. Cell cycle analysis, AO/PI staining and qPCR to assess the inducing of apoptosis as well as Tubo cancer cell inoculated mouse model for antitumor properties of STY-CFN-NPs were used. Significant increases in nanoparticle size and changes in zeta potential were observed after FA-CS coating on nanoparticles. Slow release of the STY within 144 h as well as the acceptable rate for STY encapsulation efficiency (92.4% and FA binding (52.5%) to the STY-CFN-NPs (PS: 66.26 ± 3.02 nm, ZP: 29.54 ± 1.01 mV and PDI: 0.32 ± 0.01) was reported. STY-CFN-NPs exhibited higher toxicity compared to STY suspension and treatment with STY-CFN-NPs was lead to increased apoptotic cells, stopped cells in the SubG1 phase, and also increased caspase and BAX expression and decreased BCL-2 and BCL-XL expression in in vitro and decreased the size of murine tumors (54.57% in 16 days) in in vivo. The results showed STY-CFN-NPs have good potential for breast cancer management.
Collapse
Affiliation(s)
- Soroush Sadeghi
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | | | - Amin Farhadi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
8
|
Proteomics-Based Identification of Dysregulated Proteins in Breast Cancer. Proteomes 2022; 10:proteomes10040035. [PMID: 36278695 PMCID: PMC9590004 DOI: 10.3390/proteomes10040035] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/10/2022] [Accepted: 10/18/2022] [Indexed: 11/18/2022] Open
Abstract
Immunohistochemistry (IHC) is still widely used as a morphology-based assay for in situ analysis of target proteins as specific tumor antigens. However, as a very heterogeneous collection of neoplastic diseases, breast cancer (BC) requires an accurate identification and characterization of larger panels of candidate biomarkers, beyond ER, PR, and HER2 proteins, for diagnosis and personalized treatment, without the limited availability of antibodies that are required to identify specific proteins. Top-down, middle-down, and bottom-up mass spectrometry (MS)-based proteomics approaches complement traditional histopathological tissue analysis to examine expression, modification, and interaction of hundreds to thousands of proteins simultaneously. In this review, we discuss the proteomics-based identification of dysregulated proteins in BC that are essential for the following issues: discovery and validation of new biomarkers by analysis of solid and liquid/non-invasive biopsies, cell lines, organoids and xenograft models; identification of panels of biomarkers for early detection and accurate discrimination between cancer, benign and normal tissues; identification of subtype-specific and stage-specific protein expression profiles in BC grading and measurement of disease progression; characterization of new subtypes of BC; characterization and quantitation of post-translational modifications (PTMs) and aberrant protein-protein interactions (PPI) involved in tumor development; characterization of the global remodeling of BC tissue homeostasis, diagnosis and prognostic information; and deciphering of molecular functions, biological processes and mechanisms through which the dysregulated proteins cause tumor initiation, invasion, and treatment resistance.
Collapse
|
9
|
|
10
|
Correia AR, Sampaio I, Comparetti EJ, Vieira NCS, Zucolotto V. Detecting cancer cells with a highly sensitive LbL-based biosensor. Talanta 2021; 233:122506. [PMID: 34215121 DOI: 10.1016/j.talanta.2021.122506] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 10/21/2022]
Abstract
Early diagnosis of cancer is crucial for therapeutic methods to be more effective and to decrease the mortality rate due to this disease. Current diagnostic methods include imaging techniques that require expensive equipment and specialized personnel, making it difficult to apply them to many patients. To overcome these limitations, many biosensors have been developed to monitor cancer biomarkers. Here, we report on the electrochemical biosensor for selective detection of tumor cells using a simple and low-cost methodology. Layer-by-layer (LbL) self-assembly was used to modify indium tin oxide (ITO) electrodes with alternating layers of polyallylamine hydrochloride (PAH) and folic acid (FA), which binds to overexpressed folate receptors alpha (FRα) in tumor cells. The LbL-based biosensor showed high sensitivity in detecting cervical cancer cells (HeLa cells) using cyclic voltammetry (CV) and electrochemical impedance spectroscopy (EIS). A linear dependence with the logarithm cell concentration was observed and excellent detection limits were found, 4 cells mL-1 and 19 cells mL-1 for EIS and CV measurements, respectively. The developed biosensor also presented great reproducibility (RSD = 1.7%) and repeatability (RSD = 1.8%). The selectivity was confirmed after the biosensor interaction with healthy cells (HMEC cells), which did not produce significant changes in the electrochemical signals. Furthermore, it was demonstrated that selective detection of tumor cells occurs via an interaction with FA. The LbL-based biosensor provides a simple, accurate, and cost-effective platform to be applied in the early diagnosis of cancer.
Collapse
Affiliation(s)
- Abilene Rodrigues Correia
- GNano - Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, CP 369, 13560-970, São Carlos, SP, Brazil
| | - Isabella Sampaio
- GNano - Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, CP 369, 13560-970, São Carlos, SP, Brazil.
| | - Edson José Comparetti
- GNano - Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, CP 369, 13560-970, São Carlos, SP, Brazil
| | - Nirton Cristi Silva Vieira
- GNano - Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, CP 369, 13560-970, São Carlos, SP, Brazil; Institute of Science and Technology, Federal University of São Paulo, 12231-280, São José dos Campos, SP, Brazil
| | - Valtencir Zucolotto
- GNano - Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, CP 369, 13560-970, São Carlos, SP, Brazil
| |
Collapse
|
11
|
Mansur AAP, Mansur HS, Leonel AG, Carvalho IC, Lage MCG, Carvalho SM, Krambrock K, Lobato ZIP. Supramolecular magnetonanohybrids for multimodal targeted therapy of triple-negative breast cancer cells. J Mater Chem B 2021; 8:7166-7188. [PMID: 32614035 DOI: 10.1039/d0tb01175d] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Despite the undeniable advances in recent decades, cancer remains one of the deadliest diseases of the current millennium, where the triple-negative breast cancer (TNBC) is very aggressive, extremely metastatic, and resistant to conventional chemotherapy. The nanotheranostic approach focusing on targeting membrane receptors often expressed at abnormal levels by cancer cells can be a strategic weapon for fighting malignant tumors. Herein, we introduced a novel "all-in-one nanosoldier" made of colloidal hybrid nanostructures, which were designed for simultaneously targeting, imaging, and killing TNBC cells. These nanohybrids comprised four distinct components: (a) superparamagnetic iron oxide nanoparticles, as bi-functional nanomaterials for inducing ferroptosis via inorganic nanozyme-mediated catalysis and magnetotherapy by hyperthermia treatment; (b) carboxymethyl cellulose biopolymer, as a water-soluble capping macromolecule; (c) folic acid, as the membranotopic vector for targeting folate receptors; (d) and doxorubicin (DOX) drug for chemotherapy. The results demonstrated that this novel strategy was highly effective for targeting and killing TNBC cells in vitro, expressing high levels of folate membrane-receptors. The results evidenced that three integrated mechanisms triggered the deaths of the cancer cells in vitro: (a) ferroptosis, by magnetite nanoparticles inducing a Fenton-like reaction; (b) magneto-hyperthermia effect by generating heat under an alternate magnetic field; and (c) chemotherapy, through the DOX intracellular release causing DNA dysfunction. This "all-in-one nanosoldier" strategy offers a vast realm of prospective alternatives for attacking cancer cells, combining multimodal therapy and the delivery of therapeutic agents to diseased sites and preserving healthy cells, which is one of the most critical clinical challenges faced in fighting drug-resistant breast cancers.
Collapse
Affiliation(s)
- Alexandra A P Mansur
- Center of Nanoscience, Nanotechnology and Innovation - CeNano2I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 - Escola de Engenharia, Bloco 2 - Sala 2233, 31.270-901, Belo Horizonte/M.G., Brazil.
| | - Herman S Mansur
- Center of Nanoscience, Nanotechnology and Innovation - CeNano2I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 - Escola de Engenharia, Bloco 2 - Sala 2233, 31.270-901, Belo Horizonte/M.G., Brazil.
| | - Alice G Leonel
- Center of Nanoscience, Nanotechnology and Innovation - CeNano2I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 - Escola de Engenharia, Bloco 2 - Sala 2233, 31.270-901, Belo Horizonte/M.G., Brazil.
| | - Isadora C Carvalho
- Center of Nanoscience, Nanotechnology and Innovation - CeNano2I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 - Escola de Engenharia, Bloco 2 - Sala 2233, 31.270-901, Belo Horizonte/M.G., Brazil.
| | - Manuela C G Lage
- Center of Nanoscience, Nanotechnology and Innovation - CeNano2I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 - Escola de Engenharia, Bloco 2 - Sala 2233, 31.270-901, Belo Horizonte/M.G., Brazil.
| | - Sandhra M Carvalho
- Center of Nanoscience, Nanotechnology and Innovation - CeNano2I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 - Escola de Engenharia, Bloco 2 - Sala 2233, 31.270-901, Belo Horizonte/M.G., Brazil.
| | - Klaus Krambrock
- Department of Physics, Federal University of Minas Gerais - UFMG, Brazil
| | - Zelia I P Lobato
- Department of Preventive Veterinary Medicine, Veterinary School, Federal University of Minas Gerais - UFMG, Brazil
| |
Collapse
|
12
|
Xu M, Lu M, Zhang W, Jin Q, Chen Y. Simultaneous Detection of Six Isoforms of Tau Protein in Human Cerebrospinal Fluid by Multidimensional Mass Spectrometry-Based Targeted Proteomics. J Proteome Res 2021; 20:2299-2307. [PMID: 33843226 DOI: 10.1021/acs.jproteome.0c00826] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Abnormal expression of Tau protein can cause the development of Alzheimer's disease (AD). So far, much evidence has demonstrated that Tau has multiple isoforms. These isoforms are suggested to have distinct physiological roles and contribute unequally to the progress of AD. Thus, detection of individual Tau isoforms may be helpful to better understand the link between clinical outcome and Tau status and to further improve AD diagnosis and treatment. However, few studies have been conducted on absolute quantification of Tau isoforms, probably due to high sequence homology and also low abundance of these isoforms in biofluids such as cerebrospinal fluid (CSF). Therefore, mass spectrometry-based targeted proteomics was attempted here. This targeted proteomics approach can principally measure a protein of interest at the surrogate peptide level, yet little has been done to detect protein isoforms, probably due to lack of isoform-specific surrogate peptides in mass spectrometry. In this study, separations in more dimensions were added, including immunoprecipitation (IP) and sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) for sample pretreatment and systems of linear equations for post-lab data extraction. Moreover, the reliability of the approach including IP enrichment, gel separation, and linear algebra algorithms was discussed. As a result, each isoform of Tau protein can be individually detected and quantified. Using IP enrichment, ∼250-fold enhancement of sensitivity was achieved. The ultimate LOQ was 0.50 nM. Finally, this multidimensional mass spectrometry-based targeted proteomics assay was validated and applied to simultaneous quantitative analysis of six Tau isoforms in CSF of AD patients.
Collapse
Affiliation(s)
- Mengying Xu
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Meiyan Lu
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Wenjun Zhang
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Qingwen Jin
- Sir Run Run Hospital Affiliated to Nanjing Medical University, Nanjing 211100, China
| | - Yun Chen
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.,State Key Laboratory of Reproductive Medicine, Nanjing 210029, China.,Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Nanjing 210029, China
| |
Collapse
|
13
|
Guncheva M, Idakieva K, Todinova S, Stoyanova E, Yancheva D. Folate-conjugated Helix lucorum hemocyanin - preparation, stability, and cytotoxicity. ACTA ACUST UNITED AC 2020; 75:23-30. [PMID: 31926108 DOI: 10.1515/znc-2019-0144] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 12/11/2019] [Indexed: 11/15/2022]
Abstract
This is the first report on the modification of a hemocyanin from Helix lucorum (HlH), a large molluscan respiratory protein, with folic acid (FA). In a two-step synthetic reaction, we prepared samples of HlH conjugated with 20 and 50 FA residues denoted as FA-HlH-1 and FA-HlH-2, respectively. Comparison of the attenuated total reflectance-Fourier transform infrared spectra in the amide I band region showed a structural rearrangement in the HlH that is due to FA conjugation. The changes in the secondary structure were more noticeable for FA-HlH-2. The thermal stability of HlH was not significantly affected by the FA modification, which is consistent with the observed structural similarities with the native protein. Preliminary cytotoxicity assays showed that FA-HlH-1 and FA-HlH-2 stimulate fibroblast proliferation when applied in concentrations of 50 and 100 μg/well. A negligible reduction of fibroblast growth was observed only for FA-HlH-1 and FA-HlH-2, exposed to 200 μg/well for 48 h. We found that FA-HlH-2 exhibits a low to moderate cytotoxic effect on two breast cancer cell lines, which express folate receptors, a hormone-dependent (MCF-7) and a hormone-independent (MDA-MB-231). FA-HlH-2 protects nontransformed cells and affects only neoplastic cells, which could be an advantage, and the protein could have potential in combination with other chemotherapeutics.
Collapse
Affiliation(s)
- Maya Guncheva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Krassimira Idakieva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Svetla Todinova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Elena Stoyanova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Denitsa Yancheva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Sofia, Bulgaria
| |
Collapse
|
14
|
Birlik Demirel G, Aygul E, Dag A, Atasoy S, Cimen Z, Cetin B. Folic Acid-Conjugated pH and Redox-Sensitive Ellipsoidal Hybrid Magnetic Nanoparticles for Dual-Triggered Drug Release. ACS APPLIED BIO MATERIALS 2020; 3:4949-4961. [DOI: 10.1021/acsabm.0c00488] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Gokcen Birlik Demirel
- Department of Chemistry, Polatlı Faculty of Arts and Sciences, Ankara Hacı Bayram Veli University, 06900 Ankara, Turkey
| | - Ebru Aygul
- Department of Chemistry, Polatlı Faculty of Arts and Sciences, Ankara Hacı Bayram Veli University, 06900 Ankara, Turkey
- Department of Chemistry, Institute of Natural and Applied Sciences, Gazi University, 06500 Ankara, Turkey
| | - Aydan Dag
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Bezmialem Vakif University, 34093 Istanbul, Turkey
| | - Sezen Atasoy
- Department of Biochemistry, Faculty of Pharmacy, Bezmialem Vakif University, 34093 Istanbul, Turkey
| | - Zeynep Cimen
- Department of Chemistry, Polatlı Faculty of Arts and Sciences, Ankara Hacı Bayram Veli University, 06900 Ankara, Turkey
- Department of Chemistry, Institute of Natural and Applied Sciences, Gazi University, 06500 Ankara, Turkey
| | - Busra Cetin
- Department of Chemistry, Polatlı Faculty of Arts and Sciences, Ankara Hacı Bayram Veli University, 06900 Ankara, Turkey
- Department of Chemistry, Institute of Natural and Applied Sciences, Gazi University, 06500 Ankara, Turkey
| |
Collapse
|
15
|
Omurtag Ozgen PS, Atasoy S, Zengin Kurt B, Durmus Z, Yigit G, Dag A. Glycopolymer decorated multiwalled carbon nanotubes for dual targeted breast cancer therapy. J Mater Chem B 2020; 8:3123-3137. [PMID: 32211704 DOI: 10.1039/c9tb02711d] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Carbon-based nanomaterials (CNMs) have attracted great attention in biomedical applications such as cancer imaging and therapy. CNMs, which are currently used in a wide range of applications, suffer from drawbacks of toxicity and low biocompatibility. Either noncovalent or covalent functionalization of CNMs with hydrophilic and biocompatible polymers which help to block hydrophobic interactivity between CNMs and cells can greatly increase their biocompatibility by eliminating their probable toxicity towards living organisms. In this report, we present a comparison of both noncovalent and covalent functionalization approaches in order to introduce a biocompatible glycoblock copolymer onto multi-walled carbon nanotubes (CNTs) in order to enhance their potential in therapies. An anticancer drug (doxorubicin, Dox) was conjugated with two different end functionalized poly(1-O-methacryloyl-β-d-fructopyranose-b-(2-methacryloxyethoxy))benzaldehyde glycoblock copolymers, which were synthesized via reversible addition-fragmentation chain transfer (RAFT) polymerization, by either noncovalent or covalent tethering. CNTs were coated separately with the synthesized drug-conjugated glycoblock copolymers and folic acid (FA) to obtain an efficient drug delivery platform for dual-targeting of glucose transporter protein (GLUT5) and folic acid receptors (FR) in breast cancer. A library of synthesized monomers, polymers and prepared glycoblock copolymer coated CNTs (hybrid-CNTs) using both approaches were comprehensively characterized by various techniques. Transmission electron microscopy measurements showed the homogeneous, smooth morphology of the prepared Dox-conjugated glycoblock copolymer coating of CNTs and confocal laser scanning microscopy images displayed successful cellular internalization of hybrid-CNTs in the MCF-7 and MDA-MB-231 human breast cancer cell lines. This research demonstrates the potential of hybrid-CNTs as a biocompatible drug delivery system as well as in vitro use of Dox-conjugated vehicles for dual receptor mediated breast cancer therapy.
Collapse
Affiliation(s)
- Pinar Sinem Omurtag Ozgen
- Department of Analytical Chemistry, Faculty of Pharmacy, Istanbul Medipol University, 34815, Istanbul, Turkey.
| | | | | | | | | | | |
Collapse
|
16
|
Zhang T, Zhang W, Liu L, Chen Y. Simultaneous detection of site-specific histone methylations and acetylation assisted by single template oriented molecularly imprinted polymers. Analyst 2020; 145:1376-1383. [DOI: 10.1039/c9an02360g] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A targeted proteomics assay combining single template oriented MIPs with LC-MS/MS for the simultaneous quantification of histone post-translational modification.
Collapse
Affiliation(s)
- Tianqi Zhang
- School of Pharmarcy, Nanjing Medical University
- Nanjing
- China
| | - Wen Zhang
- School of Pharmarcy, Nanjing Medical University
- Nanjing
- China
| | - Liang Liu
- School of Pharmarcy, Nanjing Medical University
- Nanjing
- China
| | - Yun Chen
- School of Pharmarcy, Nanjing Medical University
- Nanjing
- China
- State Key Laboratory of Reproductive Medicine
- China
| |
Collapse
|
17
|
Poudel BK, Hwang J, Ku SK, Kim JO, Byeon JH. Plug-and-Play Continuous Gas Flow Assembly of Cysteine-Inserted AuCu Nanobimetals for Folate-Receptor-Targeted Chemo-Phototherapy. ACS APPLIED MATERIALS & INTERFACES 2019; 11:17193-17203. [PMID: 31012571 DOI: 10.1021/acsami.9b02330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Conjugatable nanobimetals exhibiting broadband light absorption for use as phototherapeutic platforms were assembled via a plug-and-play continuous gas flow route. Electrically produced AuCu nanobunches (NBs) under nitrogen gas flow were directly injected into cysteine (cys) solution through gas pressurization to mechanically spray the solution (AuCu into cys droplets). The sprayed droplets were then exposed to 185 nm UV light (higher photon energy [6.2 eV] than the work functions of Au [5.1 eV] and Cu [4.7 eV]) to initiate photoionization of AuCu NBs for subsequent electrostatic reaction with the SH- group of cys to form cys-inserted AuCu (AuCu-cys) platforms in a single-pass gas stream. These platforms exhibited broadband light absorption spectra because of hybridized interparticle plasmonic coupling and could be conjugated to folic acid (FA) when dispersed in FA solution to form highly dispersible, biocompatible, and cancer-targetable AuCu-cys-FA. This material was suitable for use in targeted phototherapy of folate-receptor (FR)-rich cancers via FR-mediated endocytosis, and loading doxorubicin (DOX) into AuCu-cys-FA (i.e., AuCu-cys-DOXFA) facilitated chemo-phototherapy because of photoresponsive anticancer drug release upon induction of hyperthermia.
Collapse
Affiliation(s)
- Bijay Kumar Poudel
- School of Mechanical Engineering , Yonsei University , Seoul 03722 , Republic of Korea
| | - Jungho Hwang
- School of Mechanical Engineering , Yonsei University , Seoul 03722 , Republic of Korea
| | - Sae Kwang Ku
- College of Korean Medicine , Daegu Haany University , Gyeongsan 38610 , Republic of Korea
| | | | | |
Collapse
|
18
|
Abstract
Targeted proteomics detects proteins of interest with high sensitivity, quantitative accuracy, and reproducibility. In a targeted proteomics assay, surrogate peptides are generated by proteolytic digestion of target proteins and selected reaction monitoring (SRM) assays are developed to quantify these peptides using liquid chromatography-tandem mass spectrometry (LC-MS/MS). In this report, we describe the details of quantitative analysis of target protein in cells and tissue samples.
Collapse
Affiliation(s)
- Yun Chen
- School of Pharmacy, Nanjing Medical University, Nanjing, China.
| | - Liang Liu
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| |
Collapse
|
19
|
Kayani Z, Bordbar AK, Firuzi O. Novel folic acid-conjugated doxorubicin loaded β-lactoglobulin nanoparticles induce apoptosis in breast cancer cells. Biomed Pharmacother 2018; 107:945-956. [PMID: 30257407 DOI: 10.1016/j.biopha.2018.08.047] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/06/2018] [Accepted: 08/10/2018] [Indexed: 11/15/2022] Open
Abstract
Chemotherapy constitutes the main strategy in management of breast cancer (BC). Lack of specificity and high burden of adverse effects of chemotherapeutic agents remain the most important impediments to successful treatment of BC patients. Folate receptor α (FRα) could be very promising for therapeutic targeting in this type of cancer. In this study, ß-lactoglobulin nanoparticles (BNPs) conjugated with folic acid and loaded with doxorubicin (FDBNPs) were prepared. Various characterization techniques were applied to determine the size, polydispersity and doxorubicin loading of prepared FDBNPs in comparison with doxorubicin-loaded BNPs (DBNPs). The results showed that FDBNPs are 109.77 ± 2.80 nm in diameter with well dispersed and spherical shapes. The biodegradation of FDBNPs in the presence of trypsin enzyme and in PBS at different pH (4 and 7) was spectrophotometrically monitored and the results showed that the FDBNPs with encapsulation efficiency of 68.82%±1.76% could deliver doxorubicin at clinically relevant doses. Effects of DBNPs and FDBNPs against MCF-7 and MDA-MB-231, BC and triple negative BC (TNBC) cell lines, respectively, showed significant inhibition of cell proliferation as well as induction of apoptosis. Based on these findings, FDBNPs with facilitated drug release and targeted doxorubicin delivery capacities could have high therapeutic potential for BC and TNBC.
Collapse
Affiliation(s)
- Zahra Kayani
- Department of Biotechnology, Faculty of Advanced Sciences and Technologies, University of Isfahan, Isfahan, 81746-73441, Iran
| | | | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, 71345-3388, Iran.
| |
Collapse
|
20
|
|
21
|
Shan L, Zhuo X, Zhang F, Dai Y, Zhu G, Yung BC, Fan W, Zhai K, Jacobson O, Kiesewetter DO, Ma Y, Gao G, Chen X. A paclitaxel prodrug with bifunctional folate and albumin binding moieties for both passive and active targeted cancer therapy. Am J Cancer Res 2018; 8:2018-2030. [PMID: 29556370 PMCID: PMC5858514 DOI: 10.7150/thno.24382] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 02/02/2018] [Indexed: 12/21/2022] Open
Abstract
Folate receptor (FR) has proven to be a valuable target for chemotherapy using folic acid (FA) conjugates. However, FA-conjugated chemotherapeutics still have low therapeutic efficacy accompanied with side effects, resulting from complications such as short circulation half-life, limited tumor delivery, as well as high kidney accumulation. Herein, we present a novel FA-conjugated paclitaxel (PTX) prodrug which was additionally conjugated with an Evans blue (EB) derivative for albumin binding. The resulting bifunctional prodrug prolonged blood circulation, enhanced tumor accumulation, and consequently improved tumor therapeutic efficacy. Methods: Fmoc-Cys(Trt)-OH was coupled onto PTX at the 7'-OH position for further synthesis of ester prodrug FA-PTX-EB. The targeting ability was investigated using confocal microscopy and flow cytometry. The pharmacokinetics of this bifunctional compound was also studied. Meanwhile, cell viability was evaluated in normal cells and three cancer cell lines by MTT assay. In vivo therapeutic effect was tested on FR-α overexpressing MDA-MB-231 tumor model. Results: Compared with free PTX, the FA-PTX, PTX-EB and FA-PTX-EB prodrugs increased circulation half-life in mice from 2.19 to 3.82, 4.41, and 7.51 h, respectively. Pharmacokinetics studies showed that the FA-PTX-EB delivered more PTX to tumors than FA-PTX and free PTX. In vitro and in vivo studies demonstrated that FA-EB-conjugated PTX induced potent antitumor activity. Conclusion: FA-PTX-EB showed prolonged blood circulation, enhanced drug accumulation in tumors, higher therapeutic index, and lower side effects than either free PTX or monofunctional FA-PTX and EB-PTX. The results support the potential of using EB for the development of long-acting therapeutics.
Collapse
|
22
|
Jiang W, Liu L, Chen Y. Simultaneous Detection of Human C-Terminal p53 Isoforms by Single Template Molecularly Imprinted Polymers (MIPs) Coupled with Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS)-Based Targeted Proteomics. Anal Chem 2018; 90:3058-3066. [DOI: 10.1021/acs.analchem.7b02890] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Wenting Jiang
- School of Pharmacy, Nanjing Medical University, 818 Tian Yuan East Road, Nanjing, Jiangsu, China, 211166
| | - Liang Liu
- School of Pharmacy, Nanjing Medical University, 818 Tian Yuan East Road, Nanjing, Jiangsu, China, 211166
| | - Yun Chen
- School of Pharmacy, Nanjing Medical University, 818 Tian Yuan East Road, Nanjing, Jiangsu, China, 211166
- China State Key Laboratory of Reproductive Medicine, Nanjing, China 210029
| |
Collapse
|
23
|
Anirudhan T, Christa J, Binusreejayan. pH and magnetic field sensitive folic acid conjugated protein–polyelectrolyte complex for the controlled and targeted delivery of 5-fluorouracil. J IND ENG CHEM 2018. [DOI: 10.1016/j.jiec.2017.08.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
24
|
Coupling to a cancer-selective heparan-sulfate-targeted branched peptide can by-pass breast cancer cell resistance to methotrexate. Oncotarget 2017; 8:76141-76152. [PMID: 29100299 PMCID: PMC5652693 DOI: 10.18632/oncotarget.19056] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 06/18/2017] [Indexed: 12/31/2022] Open
Abstract
Cancer-selective tetra-branched peptides, named NT4, can be coupled to different functional units for cancer cell imaging or therapy. NT4 peptides specifically bind to lipoprotein receptor-related proteins (LRP) receptors and to heparan sulfate chains on membrane proteoglycans and can be efficiently internalized by cancer cells expressing these membrane targets. Since binding and internalization of NT4 peptides is mediated by specific NT4 receptors on cancer cell membranes and this may allow drug resistance produced by drug membrane transporters to be by-passed, we tested the ability of drug-armed NT4 to by-pass drug resistance in cancer cell lines. We found that MTX-conjugated NT4 allows drug resistance to be by-passed in MTX-resistant human breast cancer cells lacking expression of folate reduced carrier. NT4 peptides appear to be extremely promising cancer-selective targeting agents that can be exploited as theranostics in personalized oncological applications.
Collapse
|
25
|
Guo C, Li X, Ye M, Xu F, Yu J, Xie C, Cao X, Guo M, Yuan Y, Zheng S. Discriminating patients with early-stage breast cancer from benign lesions by detection of oxidative DNA damage biomarker in urine. Oncotarget 2017; 8:53100-53109. [PMID: 28881796 PMCID: PMC5581095 DOI: 10.18632/oncotarget.17831] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 04/12/2017] [Indexed: 12/20/2022] Open
Abstract
Breast cancer is one of the most commonly diagnosed and death-related cancers in women worldwide. Mammography is routinely used for screening and invasive examinations such as painful tissue biopsies were recommended for patients with abnormal screening outcomes. However, a considerable proportion of these cases turn out to be benign lesions. Thus, novel non-invasive approach for discriminating breast cancer from benign lesions is desirable. Herein, we applied a high-throughput ultra performance liquid chromatography-electrospray ionization tandem mass spectrometry (UPLC-ESI-MS/MS) analysis to determine the oxidative DNA damage biomarker, 8-oxo-7,8-dihydro-2′-deoxyguanosine (8-oxodG) in urine samples from 60 patients with early-stage breast cancer (stage I, II), 51 patients with benign breast diseases and 73 healthy volunteers. We demonstrated that the concentration of urinary 8-oxodG in patients with early-stage breast cancer was significantly higher not only than that in healthy controls, but also than that in patients with benign breast diseases, whereas no significant difference of urinary 8-oxodG level was observed between benign breast diseases group and healthy control group. Moreover, there was significant difference between early-stage breast cancer group and non-cancerous group which consisted of benign breast diseases patients and healthy controls. Besides, logistic regression analysis and receiver operator characteristic (ROC) curve analysis were also performed. Our findings indicate that the marked increase of 8-oxodG in urine may serve as a potential biomarker for the risk estimation, early screening and detection of breast cancer, particularly for discriminating early-stage breast cancer from benign lesions.
Collapse
Affiliation(s)
- Cheng Guo
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Xiaofen Li
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Minfeng Ye
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.,Department of Gastrointestinal Surgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, Zhejiang 312000, China
| | - Fei Xu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Jiekai Yu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Cong Xie
- College of Chemical Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Xiaoji Cao
- College of Chemical Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Mengzhe Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical College, Xuzhou, Jiangsu 221004, China
| | - Ying Yuan
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Shu Zheng
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| |
Collapse
|
26
|
Marshalek JP, Sheeran PS, Ingram P, Dayton PA, Witte RS, Matsunaga TO. Intracellular delivery and ultrasonic activation of folate receptor-targeted phase-change contrast agents in breast cancer cells in vitro. J Control Release 2016; 243:69-77. [PMID: 27686582 DOI: 10.1016/j.jconrel.2016.09.010] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 08/01/2016] [Accepted: 09/12/2016] [Indexed: 12/22/2022]
Abstract
Breast cancer is a diverse and complex disease that remains one of the leading causes of death among women. Novel, outside-of-the-box imaging and treatment methods are needed to supplement currently available technologies. In this study, we present evidence for the intracellular delivery and ultrasound-stimulated activation of folate receptor (FR)-targeted phase-change contrast agents (PCCAs) in MDA-MB-231 and MCF-7 breast cancer cells in vitro. PCCAs are lipid-coated, perfluorocarbon-filled particles formulated as nanoscale liquid droplets capable of vaporization into gaseous microbubbles for imaging or therapy. Cells were incubated with 1:1 decafluorobutane (DFB)/octafluoropropane (OFP) PCCAs for 1h, imaged via confocal microscopy, exposed to ultrasound (9MHz, MI=1.0 or 1.5), and imaged again after insonation. FR-targeted PCCAs were observed intracellularly in both cell lines, but uptake was significantly greater (p<0.001) in MDA-MB-231 cells (93.0% internalization at MI=1.0, 79.5% at MI=1.5) than MCF-7 cells (42.4% internalization at MI=1.0, 35.7% at MI=1.5). Folate incorporation increased the frequency of intracellular PCCA detection 45-fold for MDA-MB-231 cells and 7-fold for MCF-7 cells, relative to untargeted PCCAs. Intracellularly activated PCCAs ranged from 500nm to 6μm (IQR=800nm-1.5μm) with a mean diameter of 1.15±0.59 (SD) microns. The work presented herein demonstrates the feasibility of PCCA intracellular delivery and activation using breast cancer cells, illuminating a new platform toward intracellular imaging or therapeutic delivery with ultrasound.
Collapse
Affiliation(s)
| | - Paul S Sheeran
- Physical Sciences Department, Sunnybrook Research Institute, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Pier Ingram
- Department of Medical Imaging, University of Arizona, Tucson, AZ, USA
| | - Paul A Dayton
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, NC, USA
| | - Russell S Witte
- Department of Medical Imaging, University of Arizona, Tucson, AZ, USA
| | - Terry O Matsunaga
- Department of Medical Imaging, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
27
|
A targeted proteomics approach to the quantitative analysis of ERK/Bcl-2-mediated anti-apoptosis and multi-drug resistance in breast cancer. Anal Bioanal Chem 2016; 408:7491-503. [PMID: 27510278 DOI: 10.1007/s00216-016-9847-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 07/26/2016] [Accepted: 07/28/2016] [Indexed: 10/21/2022]
Abstract
Apoptosis suppression caused by overexpression of anti-apoptotic proteins is a central factor to the acquisition of multi-drug resistance (MDR) in breast cancer. As a highly conserved anti-apoptotic protein, Bcl-2 can initiate an anti-apoptosis response via an ERK1/2-mediated pathway. However, the details therein are still far from completely understood and a quantitative description of the associated proteins in the biological context may provide more insights into this process. Following our previous attempts in the quantitative analysis of MDR mechanisms, liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based targeted proteomics was continually employed here to describe ERK/Bcl-2-mediated anti-apoptosis. A targeted proteomics assay was developed and validated first for the simultaneous quantification of ERK1/2 and Bcl-2. In particular, ERK isoforms (i.e., ERK1 and ERK2) and their differential phosphorylated forms including isobaric ones were distinguished. Using this assay, differential protein levels and site-specific phosphorylation stoichiometry were observed in parental drug-sensitive MCF-7/WT cancer cells and drug-resistant MCF-7/ADR cancer cells and breast tissue samples from two groups of patients who were either suspected or diagnosed to have drug resistance. In addition, quantitative analysis of the time course of both ERK1/2 and Bcl-2 in doxorubicin (DOX)-treated MCF-7/WT cells confirmed these findings. Overall, we propose that targeted proteomics can be used generally to resolve more complex cellular events.
Collapse
|
28
|
Xu Q, Xu F, Liu L, Chen Y. Compositional Analysis of Asymmetric and Symmetric Dimethylated H3R2 Using Liquid Chromatography–Tandem Mass Spectrometry-Based Targeted Proteomics. Anal Chem 2016; 88:8441-9. [DOI: 10.1021/acs.analchem.6b00076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Qingqing Xu
- School of Pharmacy, Nanjing Medical University, 818
Tian Yuan East Road, Nanjing, 211166, China
| | - Feifei Xu
- School of Pharmacy, Nanjing Medical University, 818
Tian Yuan East Road, Nanjing, 211166, China
| | - Liang Liu
- School of Pharmacy, Nanjing Medical University, 818
Tian Yuan East Road, Nanjing, 211166, China
| | - Yun Chen
- School of Pharmacy, Nanjing Medical University, 818
Tian Yuan East Road, Nanjing, 211166, China
| |
Collapse
|