1
|
Lee MH, Thomas JL, Lin YL, Lin HY. In vitro activation of anti-cancer gene expression by delivery of CRISPR/dCas9 ribonucleoproteins to suppress glioblastoma. Int J Biol Macromol 2025; 308:142289. [PMID: 40118423 DOI: 10.1016/j.ijbiomac.2025.142289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/07/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
Cancer has been a leading cause of death for decades. While many anti-cancer drugs exist, precisely targeting malignant cells is crucial for successful tumor treatment. This targeting can be achieved by activating anti-cancer genes, which specifically destroy malignant cells. CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) therapeutics provide a promising approach for gene activation. The technology involves utilizing the denatured Cas9 (CRISPR-associated) protein conjugated with a protein activator to deliver a ribonucleoprotein (RNP) complex including guide RNA into cells for the overexpression of specific proteins. In this study, several guide RNAs targeting cancer suppressor genes were employed. These genes included tumor protein p53 (TP53), human alpha-lactalbumin made lethal to tumor cells (HAMLET), melanoma differentiation-associated gene-7 (MDA7, IL24), phorbol-12-myristate-13-acetate-induced protein 1 (PMAIP1, NOXA), pro-apoptotic WT1 regulator (PAWR, PAR4), and TNF superfamily member 10 (TNFSF10, TRAIL). The dCas9/guide RNA complexes were then adsorbed onto magnetic epitope-imprinted nanoparticles. Uppsala 87 malignant glioma (U87MG) cells and induced astrocytes (noncancerous cells) were then treated with the RNP / nanoparticles. The overexpression of MDA7 and NOXA was monitored for at least 30 days using enzyme-linked immunosorbent assay (ELISA) kits. Finally, the induced astrocytes, first activated with these anti-cancer genes, were co-cultured with U87MG cells. This resulted in a "bystander" effect: the malignant U87MG cells underwent apoptosis, while the astrocytes survived.
Collapse
Affiliation(s)
- Mei-Hwa Lee
- Department of Materials Science and Engineering, I-Shou University, Kaohsiung 84001, Taiwan
| | - James L Thomas
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM 87131, USA
| | - Yu-Ling Lin
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 81148, Taiwan
| | - Hung-Yin Lin
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 81148, Taiwan.
| |
Collapse
|
2
|
Lalhmangaihzuala S, Vanlaldinpuia K, Khiangte V, Laldinpuii Z, Liana T, Lalhriatpuia C, Pachuau Z. Therapeutic applications of carbohydrate-based compounds: a sweet solution for medical advancement. Mol Divers 2024; 28:4553-4579. [PMID: 38554170 DOI: 10.1007/s11030-024-10810-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/10/2024] [Indexed: 04/01/2024]
Abstract
Carbohydrates, one of the most abundant biomolecules found in nature, have been seen traditionally as a dietary component of foods. Recent findings, however, have unveiled their medicinal potential in the form of carbohydrates-derived drugs. Their remarkable structural diversity, high optical purity, bioavailability, low toxicity and the presence of multiple functional groups have positioned them as a valuable scaffold and an exciting frontier in contemporary therapeutics. At present, more than 170 carbohydrates-based therapeutics have been granted approval by varying regulatory agencies such as United States Food and Drug Administration (FDA), Japan Pharmaceuticals and Medical Devices Agency (PMDA), Chinese National Medical Products Administration (NMPA), and the European Medicines Agency (EMA). This article explores an overview of the fascinating potential and impact of carbohydrate-derived compounds as pharmacological agents and drug delivery vehicles.
Collapse
Affiliation(s)
- Samson Lalhmangaihzuala
- Department of Chemistry, Pachhunga University College, Mizoram University, Aizawl, Mizoram, 796001, India
- Department of Chemistry, Mizoram University, Tanhril, Aizawl, Mizoram, 796004, India
| | - Khiangte Vanlaldinpuia
- Department of Chemistry, Pachhunga University College, Mizoram University, Aizawl, Mizoram, 796001, India.
| | - Vanlalngaihawma Khiangte
- Department of Chemistry, Pachhunga University College, Mizoram University, Aizawl, Mizoram, 796001, India
- Department of Chemistry, Mizoram University, Tanhril, Aizawl, Mizoram, 796004, India
| | - Zathang Laldinpuii
- Department of Chemistry, Pachhunga University College, Mizoram University, Aizawl, Mizoram, 796001, India
- Department of Chemistry, Mizoram University, Tanhril, Aizawl, Mizoram, 796004, India
| | - Thanhming Liana
- Department of Chemistry, Pachhunga University College, Mizoram University, Aizawl, Mizoram, 796001, India
| | - Chhakchhuak Lalhriatpuia
- Department of Chemistry, Pachhunga University College, Mizoram University, Aizawl, Mizoram, 796001, India
| | - Zodinpuia Pachuau
- Department of Chemistry, Mizoram University, Tanhril, Aizawl, Mizoram, 796004, India
| |
Collapse
|
3
|
Kincses A, Vigh JP, Petrovszki D, Valkai S, Kocsis AE, Walter FR, Lin HY, Jan JS, Deli MA, Dér A. The Use of Sensors in Blood-Brain Barrier-on-a-Chip Devices: Current Practice and Future Directions. BIOSENSORS 2023; 13:bios13030357. [PMID: 36979569 PMCID: PMC10046513 DOI: 10.3390/bios13030357] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/27/2023] [Accepted: 03/02/2023] [Indexed: 06/01/2023]
Abstract
The application of lab-on-a-chip technologies in in vitro cell culturing swiftly resulted in improved models of human organs compared to static culture insert-based ones. These chip devices provide controlled cell culture environments to mimic physiological functions and properties. Models of the blood-brain barrier (BBB) especially profited from this advanced technological approach. The BBB represents the tightest endothelial barrier within the vasculature with high electric resistance and low passive permeability, providing a controlled interface between the circulation and the brain. The multi-cell type dynamic BBB-on-chip models are in demand in several fields as alternatives to expensive animal studies or static culture inserts methods. Their combination with integrated biosensors provides real-time and noninvasive monitoring of the integrity of the BBB and of the presence and concentration of agents contributing to the physiological and metabolic functions and pathologies. In this review, we describe built-in sensors to characterize BBB models via quasi-direct current and electrical impedance measurements, as well as the different types of biosensors for the detection of metabolites, drugs, or toxic agents. We also give an outlook on the future of the field, with potential combinations of existing methods and possible improvements of current techniques.
Collapse
Affiliation(s)
- András Kincses
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (A.K.); (J.P.V.); (D.P.); (S.V.); (A.E.K.); (F.R.W.)
| | - Judit P. Vigh
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (A.K.); (J.P.V.); (D.P.); (S.V.); (A.E.K.); (F.R.W.)
- Doctoral School of Biology, University of Szeged, H-6720 Szeged, Hungary
| | - Dániel Petrovszki
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (A.K.); (J.P.V.); (D.P.); (S.V.); (A.E.K.); (F.R.W.)
- Doctoral School of Multidisciplinary Medical Sciences, University of Szeged, H-6720 Szeged, Hungary
| | - Sándor Valkai
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (A.K.); (J.P.V.); (D.P.); (S.V.); (A.E.K.); (F.R.W.)
| | - Anna E. Kocsis
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (A.K.); (J.P.V.); (D.P.); (S.V.); (A.E.K.); (F.R.W.)
| | - Fruzsina R. Walter
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (A.K.); (J.P.V.); (D.P.); (S.V.); (A.E.K.); (F.R.W.)
| | - Hung-Yin Lin
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 81148, Taiwan;
| | - Jeng-Shiung Jan
- Department of Chemical Engineering, National Cheng Kung University, Tainan 70101, Taiwan;
| | - Mária A. Deli
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (A.K.); (J.P.V.); (D.P.); (S.V.); (A.E.K.); (F.R.W.)
| | - András Dér
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (A.K.); (J.P.V.); (D.P.); (S.V.); (A.E.K.); (F.R.W.)
| |
Collapse
|
4
|
Lee MH, Lin CC, Sharma PS, Thomas JL, Lin CY, Iskierko Z, Borowicz P, Lin CY, Kutner W, Yang CH, Lin HY. Peptide Selection of MMP-1 for Electrochemical Sensing with Epitope-Imprinted Poly(TPARA- co-EDOT)s. BIOSENSORS 2022; 12:bios12111018. [PMID: 36421137 PMCID: PMC9688374 DOI: 10.3390/bios12111018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/07/2022] [Accepted: 11/10/2022] [Indexed: 06/01/2023]
Abstract
Instead of molecularly imprinting a whole protein molecule, imprinting protein epitopes is gaining popularity due to cost and solubility issues. Belonging to the matrix metalloproteinase protein family, MMP-1 is an interstitial collagenase that degrades collagen and may be involved in cell migration, cell proliferation, the pro-inflammatory effect, and cancer progression. Hence, it can serve as a disease protein biomarker and thus be useful in early diagnosis. Herein, epitopes of MMP-1 were identified by screening its crystal structure. To identify possible epitopes for imprinting, MMP-1 was cleaved in silico with trypsin, pepsin at pH = 1.3, and pepsin at pH > 2.0 using Peptide Cutter, generating peptide fragments containing 8 to 12 amino acids. Five criteria were applied to select the peptides most suitable as potential epitopes for MMP-1. The triphenylamine rhodanine-3-acetic acid (TPARA) functional monomer was synthesized to form a stable pre-polymerization complex with a selected template epitope. The complexed functional monomer was then copolymerized with 3,4-ethoxylenedioxythiophene (EDOT) using potentiodynamic electropolymerization onto indium−tin−oxide (ITO) electrodes. The composition of the molecularly imprinted poly(TPARA-co-EDOT) (MIP) was optimized by maximizing the film’s electrical conductivity. Cyclic voltammetry was used to determine MMP-1 concentration in the presence of the Fe(CN)63−/Fe(CN)64− redox probe actuating the “gate effect.” A calibration curve was constructed and used to determine the usable concentration range and the limit of detection as ca. 0.001 to 10.0 pg/mL and 0.2 fg/mL MMP-1, respectively. Finally, the MMP-1 concentration in the A549 human lung (carcinoma) culture medium was measured, and this determination accuracy was confirmed using an ELISA assay.
Collapse
Affiliation(s)
- Mei-Hwa Lee
- Department of Materials Science and Engineering, I-Shou University, Kaohsiung 84001, Taiwan
| | - Cheng-Chih Lin
- Division of Pulmonary Medicine, Department of Internal Medicine, Armed-Forces Zuoying General Hospital, Kaohsiung 81342, Taiwan
| | - Piyush Sindhu Sharma
- Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warsaw, Poland
| | - James L. Thomas
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM 87131, USA
| | - Chu-Yun Lin
- Faculty of Mathematics and Natural Sciences, School of Sciences, Institute of Chemical Sciences, Cardinal Stefan Wyszynski University in Warsaw, 01-815 Warsaw, Poland
| | - Zofia Iskierko
- Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warsaw, Poland
| | - Paweł Borowicz
- Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warsaw, Poland
| | - Chien-Yu Lin
- Faculty of Mathematics and Natural Sciences, School of Sciences, Institute of Chemical Sciences, Cardinal Stefan Wyszynski University in Warsaw, 01-815 Warsaw, Poland
| | - Wlodzimierz Kutner
- Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warsaw, Poland
- Faculty of Mathematics and Natural Sciences, School of Sciences, Institute of Chemical Sciences, Cardinal Stefan Wyszynski University in Warsaw, 01-815 Warsaw, Poland
| | - Chien-Hsin Yang
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 81148, Taiwan
| | - Hung-Yin Lin
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 81148, Taiwan
| |
Collapse
|
5
|
Lee MH, Jan JS, Thomas JL, Shih YP, Li JA, Lin CY, Ooya T, Barna L, Mészáros M, Harazin A, Porkoláb G, Veszelka S, Deli MA, Lin HY. Cellular Therapy Using Epitope-Imprinted Composite Nanoparticles to Remove α-Synuclein from an In Vitro Model. Cells 2022; 11:cells11162584. [PMID: 36010659 PMCID: PMC9406856 DOI: 10.3390/cells11162584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/09/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Several degenerative disorders of the central nervous system, including Parkinson’s disease (PD), are related to the pathological aggregation of proteins. Antibodies against toxic disease proteins, such as α-synuclein (SNCA), are therefore being developed as possible therapeutics. In this work, one peptide (YVGSKTKEGVVHGVA) from SNCA was used as the epitope to construct magnetic molecularly imprinted composite nanoparticles (MMIPs). These composite nanoparticles were characterized by dynamic light scattering (DLS), high-performance liquid chromatography (HPLC), isothermal titration calorimetry (ITC), Brunauer–Emmett–Teller (BET) analysis, and superconducting quantum interference device (SQUID) analysis. Finally, the viability of brain endothelial cells that were treated with MMIPs was measured, and the extraction of SNCA from CRISPR/dCas9a-activated HEK293T cells from the in vitro model system was demonstrated for the therapeutic application of MMIPs.
Collapse
Affiliation(s)
- Mei-Hwa Lee
- Department of Materials Science and Engineering, I-Shou University, Kaohsiung 84001, Taiwan
| | - Jeng-Shiung Jan
- Department of Chemical Engineering, National Cheng Kung University, Tainan 70101, Taiwan
| | - James L. Thomas
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM 87131, USA
| | - Yuan-Pin Shih
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 81148, Taiwan
| | - Jin-An Li
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 81148, Taiwan
| | - Chien-Yu Lin
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 81148, Taiwan
| | - Tooru Ooya
- Graduate School of Engineering, Department of Chemical Science and Engineering, Kobe University, Kobe 657-8501, Japan
- Center for Advanced Medical Engineering Research & Development (CAMED), Kobe University, Kobe 657-8501, Japan
| | - Lilla Barna
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary
- Doctoral School in Biology, University of Szeged, H-6720 Szeged, Hungary
| | - Mária Mészáros
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary
| | - András Harazin
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary
| | - Gergő Porkoláb
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary
- Doctoral School in Biology, University of Szeged, H-6720 Szeged, Hungary
| | - Szilvia Veszelka
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary
| | - Maria A. Deli
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary
- Correspondence: (M.A.D.); (H.-Y.L.)
| | - Hung-Yin Lin
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 81148, Taiwan
- Correspondence: (M.A.D.); (H.-Y.L.)
| |
Collapse
|
6
|
|
7
|
Ugbaja RN, Ogungbemi K, James AS, Peter Folorunsho A, Abolade SO, Ajamikoko SO, Atayese EO, Adedeji OV. Chitosan from Crabs (Scylla serrata) Represses Hyperlipidemia-Induced Hepato-Renal Dysfunctions in Rats: Modulation of CD43 and p53 Expression. PATHOPHYSIOLOGY 2021; 28:224-237. [PMID: 35366259 PMCID: PMC8830478 DOI: 10.3390/pathophysiology28020015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 11/29/2022] Open
Abstract
Hepato-renal dysfunctions associated with hyperlipidemia necessitates a continuous search for natural remedies. This study thus evaluated the effect of dietary chitosan on diet-induced hyperlipidemia in rats. A total of 30 male Wistar rats (90 ± 10) g were randomly allotted into six (6) groups (n = 5): Normal diet, High-fat diet (HFD), and Normal diet + 5% chitosan. The three other groups received HFD, supplemented with 1%, 3%, and 5% of chitosan. The feeding lasted for 6 weeks, after which the rats were sacrificed. The liver and kidneys were harvested for analyses. Hepatic alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP) activity, and renal biomarkers (ALT, AST, urea, and creatinine) were assayed spectrophotometrically. Additionally, expression of hepatic and renal CD43 and p53 was estimated immunohistochemically. The HFD group had elevated bodyweight compared to normal which was reversed in the chitosan-supplemented groups. Hyperlipidemia caused a significant (p < 0.05) decrease in the hepatic (AST, ALT, and ALP) and renal (AST and ALT) activities, while renal urea and creatinine increased. Furthermore, the HFD group showed an elevated level of hepatic and renal CD43 while p53 expression decreased. However, groups supplemented with chitosan showed improved hepatic and renal biomarkers, as well as corrected the aberrations in the expressions of p53 and CD43. Conclusively, dietary chitosan inclusion in the diet (between 3% and 5%) could effectively improve kidney and liver functionality via abatement of inflammatory responses.
Collapse
Affiliation(s)
- Regina Ngozi Ugbaja
- Department of Biochemistry, College of Bioscience, Federal University of Agriculture, P.M.B. 2240 Abeokuta, Nigeria; (K.O.); (A.S.J.); (A.P.F.); (S.O.A.); (S.O.A.); (E.O.A.); (O.V.A.)
- Department of Chemistry/Biochemistry, Nigerian Stored Product Research Institute, P.M.B. 5044 Ibadan, Nigeria
- Correspondence: or ; Tel.: +234-(0)7066050043
| | - Kunle Ogungbemi
- Department of Biochemistry, College of Bioscience, Federal University of Agriculture, P.M.B. 2240 Abeokuta, Nigeria; (K.O.); (A.S.J.); (A.P.F.); (S.O.A.); (S.O.A.); (E.O.A.); (O.V.A.)
- Biochemistry Program, Department of Chemical Sciences, Faculty of Science, Augustine University, P.M.B. 1010 Ilara-Epe, Nigeria
| | - Adewale Segun James
- Department of Biochemistry, College of Bioscience, Federal University of Agriculture, P.M.B. 2240 Abeokuta, Nigeria; (K.O.); (A.S.J.); (A.P.F.); (S.O.A.); (S.O.A.); (E.O.A.); (O.V.A.)
| | - Ayodele Peter Folorunsho
- Department of Biochemistry, College of Bioscience, Federal University of Agriculture, P.M.B. 2240 Abeokuta, Nigeria; (K.O.); (A.S.J.); (A.P.F.); (S.O.A.); (S.O.A.); (E.O.A.); (O.V.A.)
| | - Samuel Olanrewaju Abolade
- Department of Biochemistry, College of Bioscience, Federal University of Agriculture, P.M.B. 2240 Abeokuta, Nigeria; (K.O.); (A.S.J.); (A.P.F.); (S.O.A.); (S.O.A.); (E.O.A.); (O.V.A.)
| | - Stella Onajite Ajamikoko
- Department of Biochemistry, College of Bioscience, Federal University of Agriculture, P.M.B. 2240 Abeokuta, Nigeria; (K.O.); (A.S.J.); (A.P.F.); (S.O.A.); (S.O.A.); (E.O.A.); (O.V.A.)
| | - Eniola Olapeju Atayese
- Department of Biochemistry, College of Bioscience, Federal University of Agriculture, P.M.B. 2240 Abeokuta, Nigeria; (K.O.); (A.S.J.); (A.P.F.); (S.O.A.); (S.O.A.); (E.O.A.); (O.V.A.)
| | - Omowunmi Victoria Adedeji
- Department of Biochemistry, College of Bioscience, Federal University of Agriculture, P.M.B. 2240 Abeokuta, Nigeria; (K.O.); (A.S.J.); (A.P.F.); (S.O.A.); (S.O.A.); (E.O.A.); (O.V.A.)
| |
Collapse
|
8
|
Lee MH, Lin CC, Thomas JL, Chan CK, Lin HY. Epitope recognition of magnetic peptide-imprinted chitosan composite nanoparticles for the extraction of CRISPR/dCas9a proteins from transfected cells. NANOTECHNOLOGY 2021; 32:18LT02. [PMID: 33472179 DOI: 10.1088/1361-6528/abde00] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein (Cas9) technology is a powerful method for genetic modification (and regulation) that is of great current interest. The development of new, economical methods of detecting and extracting Cas9 (and/or dCas9) from transfected cells is thus an important advance. In this work, we employed molecular imprinting, using two peptides from the Cas9 protein, to make magnetic peptide-imprinted chitosan nanoparticles. dCas9 was encoded in a plasmid which was then transfected into human embryonic kidney (HEK293T) cells. The expression of dCas9 protein was measured by using total protein kits. Finally, the imprinted nanoparticles were used to extract dCas9 from transfected cell homogenates.
Collapse
Affiliation(s)
- Mei-Hwa Lee
- Department of Materials Science and Engineering, I-Shou University, Kaohsiung 84001, Taiwan
| | - Cheng-Chih Lin
- Division of Pulmonary Medicine, Department of Internal Medicine, Armed-Forces Zuoying General Hospital, Kaohsiung 81342, Taiwan
| | - James L Thomas
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM 87131, United States of America
| | - Chih-Kai Chan
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 81148, Taiwan
| | - Hung-Yin Lin
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 81148, Taiwan
| |
Collapse
|
9
|
Lee MH, Lin CC, Thomas JL, Li JA, Lin HY. Cellular reprogramming with multigene activation by the delivery of CRISPR/dCas9 ribonucleoproteins via magnetic peptide-imprinted chitosan nanoparticles. Mater Today Bio 2021; 9:100091. [PMID: 33521619 PMCID: PMC7820544 DOI: 10.1016/j.mtbio.2020.100091] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/26/2020] [Accepted: 12/15/2020] [Indexed: 11/08/2022] Open
Abstract
Induced pluripotent stem cells are usually derived by reprogramming transcription factors (OSKM), such as octamer-binding transcription factor 4 (OCT4), (sex determining region Y)-box 2 (SOX2), Krüppel-like factor 4 (KLF4), and cellular proto-oncogene (c-Myc). However, the genomic integration of transcription factors risks the insertion of mutations into the genome of the target cells. Recently, the clustered regularly interspaced short palindromic repeat-associated protein 9 (CRISPR/Cas9) system has been used to edit genomes. In this work, dCas9-VPR (dCas9 with a gene activator, VP64-p65-Rta (VPR), fused to its c-terminus) and guide RNA (gRNA) combined to form ribonucleoproteins, which were immobilized on magnetic peptide-imprinted chitosan nanoparticles. These were then used to activate OSKM genes in human embryonic kidney (HEK) 293T cells. Four pairs of gRNAs were used for the binding site recognition to activate the OSKM genes. Transfected HEK293T cells were then prescreened for the high expression of OSKM proteins by immunohistochemistry images. The optimal gRNAs for OSKM expression were identified using quantitative real-time polymerase chain reaction and the staining of OSKM proteins. Finally, we found that the activated expression of one of the OSKM genes is up to three-fold higher than that of the other genes, enabling precise control of the cell differentiation. Molecularly imprinted polymer nanoparticles were used to deliver dCas9 ribonucleoproteins to activate OSKM genes. Two-guide RNAs for each OSKM gene were studied. Transfected HEK293T cells were then screened using immunohistochemistry. The optimal-guide RNAs for OSKM expression were also identified using qRT-PCR.
Collapse
Affiliation(s)
- Mei-Hwa Lee
- Department of Materials Science and Engineering, I-Shou University, Kaohsiung, 84001, Taiwan
| | - Cheng-Chih Lin
- Division of Pulmonary Medicine, Department of Internal Medicine, Armed-Forces Zuoying General Hospital, Kaohsiung, 81342, Taiwan
| | - James L Thomas
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Jin-An Li
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung, 81148, Taiwan
| | - Hung-Yin Lin
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung, 81148, Taiwan
| |
Collapse
|
10
|
Lee MH, Liu KH, Thomas JL, Chen JR, Lin HY. Immunotherapy of Hepatocellular Carcinoma with Magnetic PD-1 Peptide-Imprinted Polymer Nanocomposite and Natural Killer Cells. Biomolecules 2019; 9:biom9110651. [PMID: 31731492 PMCID: PMC6920774 DOI: 10.3390/biom9110651] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 10/23/2019] [Accepted: 10/23/2019] [Indexed: 12/11/2022] Open
Abstract
Programmed cell death protein 1 (PD-1) is a biomarker on the surface of cells with a role in promoting self-tolerance by suppressing the inflammatory activity of T cells. In this work, one peptide of PD-1 was used as the template for molecular imprinting to form magnetic peptide-imprinted poly(ethylene-co-vinyl alcohol) composite nanoparticles (MPIP NPs). The nanoparticles were characterized by dynamic light scattering (DLS), high-performance liquid chromatography (HPLC), Brunauer–Emmett–Teller (BET) analysis, and superconducting quantum interference device (SQUID) analysis. Natural killer 92 (NK-92) cells were added to these composite nanoparticles and then incubated with human hepatoma (HepG2) cells. The viability and the apoptosis pathway of HepG2 were then studied using cell counting kit-8 (CCK8) and quantitative real-time polymerase chain reaction (qRT-PCR), respectively. These nanoparticles were found to significantly enhance the activity of natural killer cells toward HepG2 cells by increasing the expression of nuclear factor kappa B (NF-κB), caspase 8, and especially caspase 3.
Collapse
Affiliation(s)
- Mei-Hwa Lee
- Department of Materials Science and Engineering, I-Shou University, Kaohsiung 84001, Taiwan;
| | - Kai-Hsi Liu
- Department of Internal Medicine, Division of Cardiology, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung 813, Taiwan;
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 81148, Taiwan;
| | - James L. Thomas
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM 87131, USA;
| | - Jyun-Ren Chen
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 81148, Taiwan;
| | - Hung-Yin Lin
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 81148, Taiwan;
- Correspondence: ; Tel.: +886-(7)-591-9455 or +886-(912)-178-751
| |
Collapse
|
11
|
Lee MH, Leu CC, Lin CC, Tseng YF, Lin HY, Yang CN. Gold-decorated magnetic nanoparticles modified with hairpin-shaped DNA for fluorometric discrimination of single-base mismatch DNA. Mikrochim Acta 2019; 186:80. [PMID: 30627942 DOI: 10.1007/s00604-018-3192-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 12/15/2018] [Indexed: 11/25/2022]
Abstract
The authors describe the use of gold-decorated magnetic nanoparticles (Au/MNPs) in discriminating DNA sequences with a single-base (guanine) mismatch. The Au/MNPs were characterized through dynamic light scattering, X-ray diffraction, superconducting quantum interference device, and UV/visible spectroscopy. They were then conjugated to a probe oligomer consisting of a hairpin-shaped DNA sequence carrying two signalling fluorophores: fluorescein at its 3' end and pyrene in the loop region. When interacting with the target DNA sequences, the hybridized probe-target duplex renders the pyrene signal (at excitation/emission wavelengths of 345/375 nm) either quenched or unquenched. Quenching (or nonquenching) of the pyrene fluorescence depends on the presence of a guanine (or a nonguanine) nucleotide at the designated polymorphic site. The linear range of hybridization in these Au/MNPs is from 0.1 nM to 1.0 μM of ssDNA. Conceivably, this system may serve as a single-nucleotide polymorphism probe. Graphical Abstract Schematic presentation of probe-conjugated Au/MNP preparation (upper panel) and working principle to discriminate DNA with or without single-base (guanine) mismatch sequences at the polymorphic sites (lower panel). Py denotes pyrene-hooked pyrrolocytidine; F denotes fluorescein.
Collapse
Affiliation(s)
- Mei-Hwa Lee
- Department of Materials Science and Engineering, I-Shou University, Kaohsiung, 84001, Taiwan
| | - Ching-Chich Leu
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung, 81148, Taiwan
| | - Cheng-Chih Lin
- Division of Pulmonary Medicine, Department of Internal Medicine, Armed-Forces Zuoying General Hospital, Kaohsiung, 81342, Taiwan
| | - Yu-Fan Tseng
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung, 81148, Taiwan
| | - Hung-Yin Lin
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung, 81148, Taiwan.
- Department of Chemical and Materials Engineering, National University of Kaohsiung, 700, Kaohsiung University Road, Nan-Tzu District, Kaohsiung, 811, Taiwan.
| | - Chia-Ning Yang
- Department of Life Sciences, National University of Kaohsiung, Kaohsiung, 81148, Taiwan.
- Department of Life Sciences, National University of Kaohsiung, 700, Kaohsiung University Road, Nan-Tzu District, Kaohsiung, 811, Taiwan.
| |
Collapse
|
12
|
Mu Q, Liu G, Yang D, Kou X, Cao N, Tang Y, Miao P. Ultrasensitive Detection of DNA Based on Exonuclease III-Assisted Recycling Amplification and DNAzyme Motor. Bioconjug Chem 2018; 29:3527-3531. [DOI: 10.1021/acs.bioconjchem.8b00774] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Qianhui Mu
- Bureau of Facility Support and Budget, Chinese Academy of Sciences, Beijing, 100864, P. R. China
| | - Guangxing Liu
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, P. R. China
- University of Science and Technology of China, Hefei 230026, P. R. China
| | - Dawei Yang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, P. R. China
| | - Xinyue Kou
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, P. R. China
- University of Science and Technology of China, Hefei 230026, P. R. China
| | - Ning Cao
- Bureau of Facility Support and Budget, Chinese Academy of Sciences, Beijing, 100864, P. R. China
| | - Yuguo Tang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, P. R. China
| | - Peng Miao
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, P. R. China
- University of Science and Technology of China, Hefei 230026, P. R. China
| |
Collapse
|
13
|
Li J, Cai C, Li J, Li J, Li J, Sun T, Wang L, Wu H, Yu G. Chitosan-Based Nanomaterials for Drug Delivery. Molecules 2018; 23:E2661. [PMID: 30332830 PMCID: PMC6222903 DOI: 10.3390/molecules23102661] [Citation(s) in RCA: 230] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 10/08/2018] [Accepted: 10/11/2018] [Indexed: 12/15/2022] Open
Abstract
This review discusses different forms of nanomaterials generated from chitosan and its derivatives for controlled drug delivery. Nanomaterials are drug carriers with multiple features, including target delivery triggered by environmental, pH, thermal responses, enhanced biocompatibility, and the ability to cross the blood-brain barrier. Chitosan (CS), a natural polysaccharide largely obtained from marine crustaceans, is a promising drug delivery vector for therapeutics and diagnostics, owing to its biocompatibility, biodegradability, low toxicity, and structural variability. This review describes various approaches to obtain novel CS derivatives, including their distinct advantages, as well as different forms of nanomaterials recently developed from CS. The advanced applications of CS-based nanomaterials are presented here in terms of their specific functions. Recent studies have proven that nanotechnology combined with CS and its derivatives could potentially circumvent obstacles in the transport of drugs thereby improving the drug efficacy. CS-based nanomaterials have been shown to be highly effective in targeted drug therapy.
Collapse
Affiliation(s)
- Jianghua Li
- Key Laboratory of Marine Drugs, Ministry of Education & Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
| | - Chao Cai
- Key Laboratory of Marine Drugs, Ministry of Education & Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China.
| | - Jiarui Li
- Key Laboratory of Marine Drugs, Ministry of Education & Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
| | - Jun Li
- Key Laboratory of Marine Drugs, Ministry of Education & Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
| | - Jia Li
- Key Laboratory of Marine Drugs, Ministry of Education & Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
| | - Tiantian Sun
- Key Laboratory of Marine Drugs, Ministry of Education & Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
| | - Lihao Wang
- Key Laboratory of Marine Drugs, Ministry of Education & Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
| | - Haotian Wu
- Key Laboratory of Marine Drugs, Ministry of Education & Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
| | - Guangli Yu
- Key Laboratory of Marine Drugs, Ministry of Education & Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China.
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China.
| |
Collapse
|
14
|
Lee MH, Thomas JL, Liao CL, Jurcevic S, Crnogorac-Jurcevic T, Lin HY. Epitope recognition of peptide-imprinted polymers for Regenerating protein 1 (REG1). Sep Purif Technol 2018. [DOI: 10.1016/j.seppur.2017.09.071] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
15
|
Sheng L, Jin Y, He Y, Huang Y, Yan L, Zhao R. Well-defined magnetic surface imprinted nanoparticles for selective enrichment of 2,4-dichlorophenoxyacetic acid in real samples. Talanta 2017; 174:725-732. [DOI: 10.1016/j.talanta.2017.07.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 06/22/2017] [Accepted: 07/01/2017] [Indexed: 11/26/2022]
|
16
|
Lee MH, Ahluwalia A, Chen JZ, Shih NL, Lin HY. Synthesis of magnetic cytosine-imprinted chitosan nanoparticles. NANOTECHNOLOGY 2017; 28:085705. [PMID: 28102176 DOI: 10.1088/1361-6528/aa5641] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Molecularly imprinted polymer nanoparticles incorporating magnetic nanoparticles (MNPs) have been investigated for their selective adsorption properties. Here we describe the synthesis and characterization of magnetic cytosine-imprinted chitosan nanoparticles (CIPs) for gene delivery. In particular, CIPs carrying the mammalian expression plasmid of enhanced green fluorescent protein were prepared by the co-precipitation of MNPs, chitosan and a template nucleobase (cytosine). The results show that the selective reabsorption of cytosine to magnetic CIPs was at least double that of non-imprinted polymers and other nucleobases (such as adenine and thymine). The gene carrier CIPs were used for the transfection of human embryonic kidney 293 cells showing dramatic increase their efficiency with that of conventional chitosan nanoparticles. Furthermore, the gene carrier magnetic CIPs also exhibit low toxicity compared to that of commercially available cationic lipids.
Collapse
Affiliation(s)
- Mei-Hwa Lee
- Department of Materials Science and Engineering, I-Shou University, Kaohsiung 840, Taiwan
| | | | | | | | | |
Collapse
|