1
|
Wang P, Li J, Li S, Liu Y, Gong J, He S, Wu W, Tan G, Liu S. Palladium-reduced graphene oxide nanocomposites enhance neurite outgrowth and protect neurons from Ishemic stroke. Mater Today Bio 2024; 28:101184. [PMID: 39221214 PMCID: PMC11364903 DOI: 10.1016/j.mtbio.2024.101184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/09/2024] [Accepted: 08/04/2024] [Indexed: 09/04/2024] Open
Abstract
Currently, the construction of novel biomimetic reduced graphene oxide (RGO)-based nanocomposites to induce neurite sprouting and repair the injured neurons represents a promising strategy in promoting neuronal development or treatment of cerebral anoxia or ischemia. Here, we present an effective method for constructing palladium-reduced graphene oxide (Pd-RGO) nanocomposites by covalently bonding Pd onto RGO surfaces to enhance neurite sprouting of cultured neurons. As described, the Pd-RGO nanocomposites exhibit the required physicochemical features for better biocompatibility without impacting cell viability. Primary neurons cultured on Pd-RGO nanocomposites had significantly increased number and length of neuronal processes, including both axons and dendrites, compared with the control. Western blotting showed that Pd-RGO nanocomposites improved the expression levels of growth associate protein-43 (GAP-43), as well as β-III tubulin, Tau-1, microtubule-associated protein-2 (MAP2), four proteins that are involved in regulating neurite sprouting and outgrowth. Importantly, Pd-RGO significantly promoted neurite length and complexity under oxygen-glucose deprivation/re-oxygenation (OGD/R) conditions, an in vitro cellular model of ischemic brain damage, that closely relates to neuronal GAP-43 expression. Furthermore, using the middle cerebral artery occlusion (MCAO) model in rats, we found Pd-RGO effectively reduced the infarct area, decreased neuronal apoptosis in the brain, and improved the rats' behavioral outcomes after MCAO. Together, these results indicate the great potential of Pd-RGO nanocomposites as a novel excellent biomimetic material for neural interfacing that shed light on its applications in brain injuries.
Collapse
Affiliation(s)
- Ping Wang
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Brain Science & Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Jinling Li
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Brain Science & Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Shuntang Li
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Brain Science & Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Yuanyuan Liu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Brain Science & Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Jiangu Gong
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Brain Science & Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Shipei He
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Brain Science & Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Weifeng Wu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Guohe Tan
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Brain Science & Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Sijia Liu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration &Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Brain Science & Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
2
|
Lu B, Wei L, Shi G, Du J. Nanotherapeutics for Alleviating Anesthesia-Associated Complications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308241. [PMID: 38342603 PMCID: PMC11022745 DOI: 10.1002/advs.202308241] [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: 10/30/2023] [Revised: 12/22/2023] [Indexed: 02/13/2024]
Abstract
Current management of anesthesia-associated complications falls short in terms of both efficacy and safety. Nanomaterials with versatile properties and unique nano-bio interactions hold substantial promise as therapeutics for addressing these complications. This review conducts a thorough examination of the existing nanotherapeutics and highlights the strategies for developing prospective nanomedicines to mitigate anesthetics-related toxicity. Initially, general, regional, and local anesthesia along with the commonly used anesthetics and related prevalent side effects are introduced. Furthermore, employing nanotechnology to prevent and alleviate the complications of anesthetics is systematically demonstrated from three aspects, that is, developing 1) safe nano-formulization for anesthetics; 2) nano-antidotes to sequester overdosed anesthetics and alter their pharmacokinetics; 3) nanomedicines with pharmacodynamic activities to treat anesthetics toxicity. Finally, the prospects and challenges facing the clinical translation of nanotherapeutics for anesthesia-related complications are discussed. This work provides a comprehensive roadmap for developing effective nanotherapeutics to prevent and mitigate anesthesia-associated toxicity, which can potentially revolutionize the management of anesthesia complications.
Collapse
Affiliation(s)
- Bin Lu
- Department of AnesthesiologyThird Hospital of Shanxi Medical UniversityShanxi Bethune HospitalShanxi Academy of Medical SciencesTongji Shanxi HospitalTaiyuan030032China
- Key Laboratory of Cellular Physiology at Shanxi Medical UniversityMinistry of EducationTaiyuanShanxi Province030001China
| | - Ling Wei
- Shanxi Bethune Hospital Center Surgery DepartmentShanxi Academy of Medical SciencesTongji Shanxi HospitalThird Hospital of Shanxi Medical UniversityTaiyuan030032China
| | - Gaoxiang Shi
- Department of AnesthesiologyThird Hospital of Shanxi Medical UniversityShanxi Bethune HospitalShanxi Academy of Medical SciencesTongji Shanxi HospitalTaiyuan030032China
| | - Jiangfeng Du
- Key Laboratory of Cellular Physiology at Shanxi Medical UniversityMinistry of EducationTaiyuanShanxi Province030001China
- Department of Medical ImagingShanxi Key Laboratory of Intelligent Imaging and NanomedicineFirst Hospital of Shanxi Medical UniversityTaiyuanShanxi Province030001China
| |
Collapse
|
3
|
Beltran AS. Novel Approaches to Studying SLC13A5 Disease. Metabolites 2024; 14:84. [PMID: 38392976 PMCID: PMC10890222 DOI: 10.3390/metabo14020084] [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: 11/18/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 02/25/2024] Open
Abstract
The role of the sodium citrate transporter (NaCT) SLC13A5 is multifaceted and context-dependent. While aberrant dysfunction leads to neonatal epilepsy, its therapeutic inhibition protects against metabolic disease. Notably, insights regarding the cellular and molecular mechanisms underlying these phenomena are limited due to the intricacy and complexity of the latent human physiology, which is poorly captured by existing animal models. This review explores innovative technologies aimed at bridging such a knowledge gap. First, I provide an overview of SLC13A5 variants in the context of human disease and the specific cell types where the expression of the transporter has been observed. Next, I discuss current technologies for generating patient-specific induced pluripotent stem cells (iPSCs) and their inherent advantages and limitations, followed by a summary of the methods for differentiating iPSCs into neurons, hepatocytes, and organoids. Finally, I explore the relevance of these cellular models as platforms for delving into the intricate molecular and cellular mechanisms underlying SLC13A5-related disorders.
Collapse
Affiliation(s)
- Adriana S Beltran
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
4
|
Ho YS, Cheng TC, Guo P. Targeted Delivery of Potent Chemical Drugs and RNAi to Drug-Resistant Breast Cancer Using RNA-Nanotechnology and RNA-Ligand Displaying Extracellular vesicles. RNA NANOMED 2024; 1:16-43. [PMID: 40125243 PMCID: PMC11927007 DOI: 10.59566/isrnn.2024.0101016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
This review describes a new technology to treat breast-cancer-drug-resistance by targeting the ABC as the multi-homo-subunit ATPase, enlightening by the Christmas-lighting budge with serial circuit and the asymmetrical homo-hexamer of the phi29 DNA packaging motor with sequential revolving mechanism. Chemotherapeutics has been widely used in breast cancer treatments, but drug resistance has raised a serious concern. RNA therapeutics has emerged as the third milestone in pharmaceutical drug development. RNA nanoparticles are dynamic, mild, and deformative, resulting in spontaneous, rapid, and efficient accumulation in tumor vasculature after IV injection. Their negative charge and favorable size bypass the nonspecific targeting of vital organs and normal cells. This motile and deformable nature also led to the fast passing of glomerular filters and their movement into the urine for rapid body clearance for those non-tumor-accumulated nanoparticles, resulting in undetectable toxicity. Extracellular vesicles have shown potential as a delivery system for RNAi and chemotherapeutic drugs in vivo, contributing to the efficacy of cancer remission. However, the lack of cell-targeting ligands on extracellular vesicles and the nonspecific entry into healthy cells has led to safety concerns. This review addresses how to apply RNA nanotechnology and RNA-ligand displaying extracellular vesicles for specific delivery to breast cancer. The particular focus is on using and combining the RNA and extracellular vesicle technology to deal with breast cancer drug resistance. The targeting capabilities and drug safety can be improved through extracellular vesicle engineering techniques, such as affixing ligands on the extracellular vesicle surface utilizing arrow-tail RNA nanoparticles, ultimately addressing off-target effects and toxicity. Using RNA ligands for specific targeting and the efficient membrane fusion of extracellular vesicles has enabled the development of ligand-displayed extracellular vesicles capable of delivering both RNAi and chemical drugs to cells with precision, effectively inhibiting tumor growth. The negative charge inherent in the vesicles results in electrostatic repulsion, reducing non-specific binding to healthy cells that contain negatively charged lipid membranes. By leveraging the principles of RNA nanotechnology, the engineering of extracellular vesicles offers a promising avenue for addressing breast cancer drug resistance. This review also discusses applying the series of circuit mechanisms in Christmas-decorating-lighting to develop effective therapeutics to combat breast cancer chemoresistance by targeting the ABC drug transporter and breast cancer surface receptors.
Collapse
Affiliation(s)
- Yuan Soon Ho
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, China Medical University, Taichung, Taiwan
| | - Tzu-Chun Cheng
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, China Medical University, Taichung, Taiwan
| | - Peixuan Guo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy; Center for RNA Nanotechnology and Nanomedicine; James Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| |
Collapse
|
5
|
Ghosh S, Dhiman M, Gupta S, Roy P, Lahiri D. Electro-conductive chitosan/graphene bio-nanocomposite scaffold for tissue engineering of the central nervous system. BIOMATERIALS ADVANCES 2023; 154:213596. [PMID: 37672898 DOI: 10.1016/j.bioadv.2023.213596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 08/04/2023] [Accepted: 08/23/2023] [Indexed: 09/08/2023]
Abstract
Degenerative central nervous system (CNS) disorders and traumatic brain injuries are common nowadays. These may induce the loss of neuronal cells and delicate connections essential for optimal CNS function. The CNS tissue has restricted regeneration ability, hindering the development of effective therapies. Developing cell and tissue instructive materials may bring up new treatment possibilities. In this study, chitosan-graphene nano platelets (GNPs) composite films were developed to regenerate brain cells. This study evaluates the effects of GNP concentration (0.5, 1 and 2 wt%) and their alignment on mechanical, electrical, surface, protein adsorption and biological properties of the regenerative scaffolds. Incorporating and aligning GNPs into chitosan matrix improved all the physical and biological properties. On reinforced scaffolds, HT22 cell morphology mimics pyramidal brain cells, which are responsible for the brain's highly branched neural network. Additionally, the reinforced scaffolds supported Mesenchymal Stem like Cells growth and were biocompatible in vivo. The alignment of GNPs in the chitosan matrix offered the appropriate physicochemical and biological properties to promote adhesion, proliferation and shape morphogenesis of hippocampal HT22 neuronal cells. Overall, this study delineates the enormous potential offered by the GNP-reinforced scaffolds for regeneration of central nervous system, especially the brain.
Collapse
Affiliation(s)
- Souvik Ghosh
- Biomaterials and Multiscale Mechanics Lab, Department of Metallurgical and Materials Engineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India; Molecular Endocrinology Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India; Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Megha Dhiman
- Biomaterials and Multiscale Mechanics Lab, Department of Metallurgical and Materials Engineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India; Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Sumeet Gupta
- Department of Pharmacy, Maharshi Markandeshwar University (Deemed to Be University), Mullana, Haryana 133207, India
| | - Partha Roy
- Molecular Endocrinology Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Debrupa Lahiri
- Biomaterials and Multiscale Mechanics Lab, Department of Metallurgical and Materials Engineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India; Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India.
| |
Collapse
|
6
|
Xu P, Li TT, Wang BC, Yi YJ, Zhang WC, Sun GD, Zhang Y, Li ZZ. Supramolecular assemblies with spatio-temporal sequential drug delivery capability treat spinal cord injury via neuroprotection and immunoregulation. J Control Release 2023; 360:528-548. [PMID: 37433370 DOI: 10.1016/j.jconrel.2023.07.008] [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: 02/02/2023] [Revised: 06/25/2023] [Accepted: 07/05/2023] [Indexed: 07/13/2023]
Abstract
Spinal cord injury (SCI) can result in irreversible motor and sensory deficits. However, up to data, clinical first-line drugs have ambiguous benefits and debilitating side effects, mainly due to the insufficient accumulation, poor physiological barrier penetration, and lack of spatio-temporal controlled release at lesion tissue. Herein, we proposed a supramolecular assemblies composed of hyperbranched polymer-formed core/shell structure through host-guest interactions. Such HPAA-BM@CD-HPG-C assemblies co-loaded with p38 inhibitor (SB203580) and insulin-like growth factor 1(IGF-1) are able to achieve time- and space-programmed sequential delivery benefiting from their cascaded responsiveness. The core-shell disassembly of HPAA-BM@CD-HPG-C occurs in acidic micro-environment around lesion, achieving preferentially the burst release of IGF-1 to protect survival neurons. Subsequently, the HPAA-BM cores containing SB203580 are endocytosed by the recruited macrophages and degraded by intracellular GSH, accelerating the release of SB203580 to promote the conversion from M1 to M2 macrophage. Hence, the successive synergy of neuroprotection and immunoregulation effects contribute to subsequent nerve repair and locomotor recovery as demonstrated in vitro and in vivo studies. Thus, our fabrication provides a strategy that multiple drugs co-delivery in a spatio-temporal selective manner adapting to the disease progression through self-cascaded disintegration, are expected to realize multidimensional precise treatment of SCI.
Collapse
Affiliation(s)
- Ping Xu
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, 601 West Whampoa Avenue, Guangzhou 510000, China
| | - Tian-Tian Li
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, 601 West Whampoa Avenue, Guangzhou 510632, China
| | - Bin-Chen Wang
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, 601 West Whampoa Avenue, Guangzhou 510632, China
| | - Yong-Jun Yi
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, 601 West Whampoa Avenue, Guangzhou 510000, China
| | - Wen-Cai Zhang
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, 601 West Whampoa Avenue, Guangzhou 510000, China
| | - Guo-Dong Sun
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, 601 West Whampoa Avenue, Guangzhou 510000, China; Key Laboratory of Guangdong Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital of Jinan University, Yingke Avenue, Heyuan City 517000, China.
| | - Yi Zhang
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, 601 West Whampoa Avenue, Guangzhou 510632, China.
| | - Zhi-Zhong Li
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, 601 West Whampoa Avenue, Guangzhou 510000, China; Key Laboratory of Guangdong Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital of Jinan University, Yingke Avenue, Heyuan City 517000, China.
| |
Collapse
|
7
|
Huang Q, Zhu W, Gao X, Liu X, Zhang Z, Xing B. Nanoparticles-mediated ion channels manipulation: From their membrane interactions to bioapplications. Adv Drug Deliv Rev 2023; 195:114763. [PMID: 36841331 DOI: 10.1016/j.addr.2023.114763] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/14/2023] [Accepted: 02/18/2023] [Indexed: 02/26/2023]
Abstract
Ion channels are transmembrane proteins ubiquitously expressed in all cells that control various ions (e.g. Na+, K+, Ca2+ and Cl- etc) crossing cellular plasma membrane, which play critical roles in physiological processes including regulating signal transduction, cell proliferation as well as excitatory cell excitation and conduction. Abnormal ion channel function is usually associated with dysfunctions and many diseases, such as neurodegenerative disorders, ophthalmic diseases, pulmonary diseases and even cancers. The precise regulation of ion channels not only helps to decipher physiological and pathological processes, but also is expected to become cutting-edge means for disease treatment. Recently, nanoparticles-mediated ion channel manipulation emerges as a highly promising way to meet the increasing requirements with respect to their simple, efficient, precise, spatiotemporally controllable and non-invasive regulation in biomedicine and other research frontiers. Thanks the advantages of their unique properties, nanoparticles can not only directly block the pore sites or kinetics of ion channels through their tiny size effect, and perturb active voltage-gated ion channel by their charged surface, but they can also act as antennas to conduct or enhance external physical stimuli to achieve spatiotemporal, precise and efficient regulation of various ion channel activities (e.g. light-, mechanical-, and temperature-gated ion channels etc). So far, nanoparticles-mediated ion channel regulation has shown potential prospects in many biomedical fields at the interfaces of neuro- and cardiovascular modulation, physiological function regeneration and tumor therapy et al. Towards such important fields, in this typical review, we specifically outline the latest studies of different types of ion channels and their activities relevant to the diseases. In addition, the different types of stimulation responsive nanoparticles, their interaction modes and targeting strategies towards the plasma membrane ion channels will be systematically summarized. More importantly, the ion channel regulatory methods mediated by functional nanoparticles and their bioapplications associated with physiological modulation and therapeutic development will be discussed. Last but not least, current challenges and future perspectives in this field will be covered as well.
Collapse
Affiliation(s)
- Qiwen Huang
- Department of Chemistry, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Weisheng Zhu
- Department of Chemistry, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Xiaoyin Gao
- Department of Chemistry, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Xinping Liu
- School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Zhijun Zhang
- Department of Chemistry, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Bengang Xing
- School of Chemistry, Chemical Engineering & Biotechnology, Nanyang Technological University, Singapore, 637371, Singapore.
| |
Collapse
|
8
|
Li J, Zhang Y, Lou Z, Li M, Cui L, Yang Z, Zhang L, Zhang Y, Gu N, Yang F. Magnetic Nanobubble Mechanical Stress Induces the Piezo1-Ca 2+ -BMP2/Smad Pathway to Modulate Neural Stem Cell Fate and MRI/Ultrasound Dual Imaging Surveillance for Ischemic Stroke. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2201123. [PMID: 35555970 DOI: 10.1002/smll.202201123] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/21/2022] [Indexed: 06/15/2023]
Abstract
Neural stem cells (NSCs) are used to treat various nervous system diseases because of their self-renewal ability and multidirectional differentiation potential. However, an insufficient ability to track their migration in vivo and poor control over their survival and differentiation efficiency are two major critical challenges for clinical application. Here, it is shown that when magnetic nanobubbles (MNBs), which are assembled from magnetic nanoparticles, are internalized by NSCs, intramembrane volumetric oscillation of the MNBs induces an increase in intracellular hydrostatic pressure and cytoskeleton force, resulting in the activation of the Piezo1-Ca2+ mechanosensory channel. This subsequently triggers the BMP2/Smad biochemical signaling pathway, leading to differentiation of NSCs into the neuronal phenotype. Signaling through the Piezo1-Ca2+ -BMP2/Smad pathway can be further accelerated by application of an external shear stress force using low-intensity pulsed ultrasound. More importantly, magnetic resonance imaging and ultrasound imaging surveillance of NSCs based on MNB labeling can be leveraged to provide NSC therapeutic outcomes. Both the in vitro and in vivo findings demonstrate that a bubble nanostructure-induced physical force can modulate and control the mechanical signaling pathway regulating stem cell development.
Collapse
Affiliation(s)
- Jing Li
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Yao Zhang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
- College of Materials Science and Engineering, Nanjing Forestry University, Nanjing, 210037, P. R. China
| | - Zhichao Lou
- College of Materials Science and Engineering, Nanjing Forestry University, Nanjing, 210037, P. R. China
| | - Mingxi Li
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing, 210009, P. R. China
| | - Lin Cui
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Zhenrong Yang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Lijuan Zhang
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201204, P. R. China
| | - Yu Zhang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
- College of Materials Science and Engineering, Nanjing Forestry University, Nanjing, 210037, P. R. China
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Fang Yang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| |
Collapse
|
9
|
Zare I, Yaraki MT, Speranza G, Najafabadi AH, Haghighi AS, Nik AB, Manshian BB, Saraiva C, Soenen SJ, Kogan MJ, Lee JW, Apollo NV, Bernardino L, Araya E, Mayer D, Mao G, Hamblin MR. Gold nanostructures: synthesis, properties, and neurological applications. Chem Soc Rev 2022; 51:2601-2680. [PMID: 35234776 DOI: 10.1039/d1cs01111a] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent advances in technology are expected to increase our current understanding of neuroscience. Nanotechnology and nanomaterials can alter and control neural functionality in both in vitro and in vivo experimental setups. The intersection between neuroscience and nanoscience may generate long-term neural interfaces adapted at the molecular level. Owing to their intrinsic physicochemical characteristics, gold nanostructures (GNSs) have received much attention in neuroscience, especially for combined diagnostic and therapeutic (theragnostic) purposes. GNSs have been successfully employed to stimulate and monitor neurophysiological signals. Hence, GNSs could provide a promising solution for the regeneration and recovery of neural tissue, novel neuroprotective strategies, and integrated implantable materials. This review covers the broad range of neurological applications of GNS-based materials to improve clinical diagnosis and therapy. Sub-topics include neurotoxicity, targeted delivery of therapeutics to the central nervous system (CNS), neurochemical sensing, neuromodulation, neuroimaging, neurotherapy, tissue engineering, and neural regeneration. It focuses on core concepts of GNSs in neurology, to circumvent the limitations and significant obstacles of innovative approaches in neurobiology and neurochemistry, including theragnostics. We will discuss recent advances in the use of GNSs to overcome current bottlenecks and tackle technical and conceptual challenges.
Collapse
Affiliation(s)
- Iman Zare
- Research and Development Department, Sina Medical Biochemistry Technologies Co. Ltd., Shiraz 7178795844, Iran
| | | | - Giorgio Speranza
- CMM - FBK, v. Sommarive 18, 38123 Trento, Italy.,IFN - CNR, CSMFO Lab., via alla Cascata 56/C Povo, 38123 Trento, Italy.,Department of Industrial Engineering, University of Trento, v. Sommarive 9, 38123 Trento, Italy
| | - Alireza Hassani Najafabadi
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA.,Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Alireza Shourangiz Haghighi
- Department of Mechanical Engineering, Shiraz University of Technology, Modarres Boulevard, 13876-71557, Shiraz, Iran
| | - Amirala Bakhshian Nik
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174, USA
| | - Bella B Manshian
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Cláudia Saraiva
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 Avenue des Hauts-Fourneaux, 4362 Esch-sur-Alzette, Luxembourg.,Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Rua Marques d'Avila e Bolama, 6201-001 Covilha, Portugal
| | - Stefaan J Soenen
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Marcelo J Kogan
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas, Departamento de Química Farmacológica y Toxicológica, Universidad de Chile, 8380492 Santiago, Chile
| | - Jee Woong Lee
- Department of Medical Sciences, Clinical Neurophysiology, Uppsala University, Uppsala, SE-751 23, Sweden
| | - Nicholas V Apollo
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,School of Physics, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Liliana Bernardino
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Rua Marques d'Avila e Bolama, 6201-001 Covilha, Portugal
| | - Eyleen Araya
- Departamento de Ciencias Quimicas, Facultad de Ciencias Exactas, Universidad Andres Bello, Av. Republica 275, Santiago, Chile
| | - Dirk Mayer
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, Germany
| | - Guangzhao Mao
- School of Chemical Engineering, University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
| | - Michael R Hamblin
- Laser Research Center, University of Johannesburg, Doorfontein 2028, South Africa.
| |
Collapse
|
10
|
Jaragh-Alhadad LA, Falahati M. Tin oxide nanoparticles trigger the formation of amyloid β oligomers/protofibrils and underlying neurotoxicity as a marker of Alzheimer's diseases. Int J Biol Macromol 2022; 204:154-160. [PMID: 35124024 DOI: 10.1016/j.ijbiomac.2022.01.190] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/11/2022] [Accepted: 01/29/2022] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is known as one of the most common forms of dementia, and oligomerization of amyloid β (Aβ42) peptides can result in the onset of AD. Tin oxide nanoparticles (SnO2 NPs) showed several applications in biomedical fields can trigger unwanted interaction with proteins and inducing protein aggregation. Herein, we synthesized SnO2 NPs via the hydrothermal method and characterized by UV-visible, XRD, FTIR, TEM, and DLS techniques. Afterward, the formation of Aβ42 amyloid oligomers/protofibrils treated alone and with SnO2 NPs was explored by ThT and Nile red fluorescence and CD spectroscopic methods along with TEM imaging. The neurotoxicity of different spices of Aβ42 samples against PC-12 cells was then explored by MTT and caspase-3 activity assays. The characterization of SnO2 NPs confirmed the successful synthesis of crystalline NPs (20-30 nm). Different biophysical and cellular analyses indicated that SnO2 NPs accelerated Aβ42 fibrillogenesis and promoted amyloid oligomers/protofibrils cytotoxicity. As compared to the Aβ42 samples grown alone, the ThT and ANS fluorescence intensity along with ellipticity results indicated the promotory effect of SnO2 NPs on the formation of oligomers/protofibrils. Also, the cellular results showed that the treated Aβ42 samples with SnO2 NPs further reduced cell viability through activation of caspase-3. In conclusion, SnO2 NPs greatly accelerate the fibrillation of Aβ42 peptides and lead to the formation of more toxic species. The present data may offer further warrants into nano-based systems for biomedical applications in the central nervous system.
Collapse
Affiliation(s)
- Laila Abdulmohsen Jaragh-Alhadad
- Department of Chemistry, College of Science, Kuwait University, Safat 13060, Kuwait; Cardiovascular and Metabolic Sciences Department, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio 44195, USA.
| | - Mojtaba Falahati
- Laboratory Experimental Oncology and Nanomedicine Innovation Center Erasmus (NICE), Department of Pathology, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Nanotechnology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
11
|
Yu D, Wang J, Qian KJ, Yu J, Zhu HY. Effects of nanofibers on mesenchymal stem cells: environmental factors affecting cell adhesion and osteogenic differentiation and their mechanisms. J Zhejiang Univ Sci B 2021; 21:871-884. [PMID: 33150771 DOI: 10.1631/jzus.b2000355] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Nanofibers can mimic natural tissue structure by creating a more suitable environment for cells to grow, prompting a wide application of nanofiber materials. In this review, we include relevant studies and characterize the effect of nanofibers on mesenchymal stem cells, as well as factors that affect cell adhesion and osteogenic differentiation. We hypothesize that the process of bone regeneration in vitro is similar to bone formation and healing in vivo, and the closer nanofibers or nanofibrous scaffolds are to natural bone tissue, the better the bone regeneration process will be. In general, cells cultured on nanofibers have a similar gene expression pattern and osteogenic behavior as cells induced by osteogenic supplements in vitro. Genes involved in cell adhesion (focal adhesion kinase (FAK)), cytoskeletal organization, and osteogenic pathways (transforming growth factor-β (TGF-β)/bone morphogenic protein (BMP), mitogen-activated protein kinase (MAPK), and Wnt) are upregulated successively. Cell adhesion and osteogenesis may be influenced by several factors. Nanofibers possess certain physical properties including favorable hydrophilicity, porosity, and swelling properties that promote cell adhesion and growth. Moreover, nanofiber stiffness plays a vital role in cell fate, as cell recruitment for osteogenesis tends to be better on stiffer scaffolds, with associated signaling pathways of integrin and Yes-associated protein (YAP)/transcriptional co-activator with PDZ-binding motif (TAZ). Also, hierarchically aligned nanofibers, as well as their combination with functional additives (growth factors, HA particles, etc.), contribute to osteogenesis and bone regeneration. In summary, previous studies have indicated that upon sensing the stiffness of the nanofibrous environment as well as its other characteristics, stem cells change their shape and tension accordingly, regulating downstream pathways followed by adhesion to nanofibers to contribute to osteogenesis. However, additional experiments are needed to identify major signaling pathways in the bone regeneration process, and also to fully investigate its supportive role in fabricating or designing the optimum tissue-mimicking nanofibrous scaffolds.
Collapse
Affiliation(s)
- Dan Yu
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jin Wang
- Department of Stomatology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Ke-Jia Qian
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jing Yu
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Hui-Yong Zhu
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
12
|
Halim A, Qu KY, Zhang XF, Huang NP. Recent Advances in the Application of Two-Dimensional Nanomaterials for Neural Tissue Engineering and Regeneration. ACS Biomater Sci Eng 2021; 7:3503-3529. [PMID: 34291638 DOI: 10.1021/acsbiomaterials.1c00490] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The complexity of the nervous system structure and function, and its slow regeneration rate, makes it more difficult to treat compared to other tissues in the human body when an injury occurs. Moreover, the current therapeutic approaches including the use of autografts, allografts, and pharmacological agents have several drawbacks and can not fully restore nervous system injuries. Recently, nanotechnology and tissue engineering approaches have attracted many researchers to guide tissue regeneration in an effective manner. Owing to their remarkable physicochemical and biological properties, two-dimensional (2D) nanomaterials have been extensively studied in the tissue engineering and regenerative medicine field. The great conductivity of these materials makes them a promising candidate for the development of novel scaffolds for neural tissue engineering application. Moreover, the high loading capacity of 2D nanomaterials also has attracted many researchers to utilize them as a drug/gene delivery method to treat various devastating nervous system disorders. This review will first introduce the fundamental physicochemical properties of 2D nanomaterials used in biomedicine and the supporting biological properties of 2D nanomaterials for inducing neuroregeneration, including their biocompatibility on neural cells, the ability to promote the neural differentiation of stem cells, and their immunomodulatory properties which are beneficial for alleviating chronic inflammation at the site of the nervous system injury. It also discusses various types of 2D nanomaterials-based scaffolds for neural tissue engineering applications. Then, the latest progress on the use of 2D nanomaterials for nervous system disorder treatment is summarized. Finally, a discussion of the challenges and prospects of 2D nanomaterials-based applications in neural tissue engineering is provided.
Collapse
Affiliation(s)
- Alexander Halim
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P.R. China
| | - Kai-Yun Qu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P.R. China
| | - Xiao-Feng Zhang
- Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, P.R. China
| | - Ning-Ping Huang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P.R. China
| |
Collapse
|
13
|
Patel M, Jha A, Patel R. Potential application of PLGA microsphere for tissue engineering. JOURNAL OF POLYMER RESEARCH 2021. [DOI: 10.1007/s10965-021-02562-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
14
|
Liu L, Wu J, Wang S, Kun L, Gao J, Chen B, Ye Y, Wang F, Tong F, Jiang J, Ou J, Wilson DA, Tu Y, Peng F. Control the Neural Stem Cell Fate with Biohybrid Piezoelectrical Magnetite Micromotors. NANO LETTERS 2021; 21:3518-3526. [PMID: 33848170 DOI: 10.1021/acs.nanolett.1c00290] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Inducing neural stem cells to differentiate and replace degenerated functional neurons represents the most promising approach for neural degenerative diseases including Parkinson's disease, Alzheimer's disease, etc. While diverse strategies have been proposed in recent years, most of these are hindered due to uncontrollable cell fate and device invasiveness. Here, we report a minimally invasive micromotor platform with biodegradable helical Spirulina plantensis (S. platensis) as the framework and superparamagnetic Fe3O4 nanoparticles/piezoelectric BaTiO3 nanoparticles as the built-in function units. With a low-strength rotational magnetic field, this integrated micromotor system can perform precise navigation in biofluid and achieve single-neural stem cell targeting. Remarkably, by tuning ultrasound intensity, thus the local electrical output by the motor, directed differentiation of the neural stem cell into astrocytes, functional neurons (dopamine neurons, cholinergic neurons), and oligodendrocytes, can be achieved. This micromotor platform can serve as a highly controllable wireless tool for bioelectronics and neuronal regenerative therapy.
Collapse
Affiliation(s)
- Lu Liu
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
- School of Pharmaceutical Science, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Juanyan Wu
- School of Pharmaceutical Science, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Shuanghu Wang
- School of Pharmaceutical Science, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Liu Kun
- School of Pharmaceutical Science, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Junbin Gao
- School of Pharmaceutical Science, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Bin Chen
- School of Pharmaceutical Science, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Yicheng Ye
- School of Pharmaceutical Science, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Fei Wang
- School of Pharmaceutical Science, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Fei Tong
- School of Pharmaceutical Science, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Jiamiao Jiang
- School of Pharmaceutical Science, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Juanfeng Ou
- School of Pharmaceutical Science, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Daniela A Wilson
- Institute for Molecules and Materials, Radboud University, Nijmegen 6525 AJ, The Netherlands
| | - Yingfeng Tu
- School of Pharmaceutical Science, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Fei Peng
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| |
Collapse
|
15
|
Wei M, Li S, Yang Z, Cheng C, Li T, Le W. Tetrahedral DNA nanostructures functionalized by multivalent microRNA132 antisense oligonucleotides promote the differentiation of mouse embryonic stem cells into dopaminergic neurons. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 34:102375. [PMID: 33617970 DOI: 10.1016/j.nano.2021.102375] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 11/29/2022]
Abstract
MicroRNA132 (miR132) negatively regulates the differentiation of mouse embryonic stem cells (ESCs) into dopaminergic (DAergic) neurons; in contrast, antisense oligonucleotide against miR132 (miR132-ASO) effectively blocks the activity of endogenous miR132 and thereafter promotes the differentiation of DAergic neurons. However, it is difficult for miR132-ASO to enter cells without a suitable delivery system. Tetrahedral DNA nanostructures (TDNs), as a new type of DNA-based nanocarrier, have great potential in biomedical applications and even have been reported to promote stem cell differentiation. In this study, we developed functional multivalent DNA nanostructures by appending miR132-ASO motifs to three-dimensional TDNs (miR132-ASO-TDNs). Our data clearly revealed that miR132-ASO-TDNs exposure can promote the differentiation of ESCs into DAergic neurons as well as elevate DA release from differentiated DAergic neurons. MiR132-ASO-TDNs could serve as a novel biofunctional nanomaterial to improve the efficiency of DAergic neurons differentiation. Our findings may also provide a new approach for stem cell therapy against neurodegenerative diseases.
Collapse
Affiliation(s)
- Min Wei
- Liaoning Provincial Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, People's Republic of China; Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, People's Republic of China
| | - Song Li
- Liaoning Provincial Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, People's Republic of China; Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, People's Republic of China
| | - Zhaofei Yang
- Liaoning Provincial Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, People's Republic of China; Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, People's Republic of China
| | - Cheng Cheng
- Liaoning Provincial Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, People's Republic of China; Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, People's Republic of China
| | - Tianbai Li
- Liaoning Provincial Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, People's Republic of China; Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, People's Republic of China
| | - Weidong Le
- Liaoning Provincial Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, People's Republic of China; Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, People's Republic of China.
| |
Collapse
|
16
|
Lee HP, Gaharwar AK. Light-Responsive Inorganic Biomaterials for Biomedical Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2000863. [PMID: 32995121 PMCID: PMC7507067 DOI: 10.1002/advs.202000863] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/24/2020] [Indexed: 05/19/2023]
Abstract
Light-responsive inorganic biomaterials are an emerging class of materials used for developing noninvasive, noncontact, precise, and controllable medical devices in a wide range of biomedical applications, including photothermal therapy, photodynamic therapy, drug delivery, and regenerative medicine. Herein, a range of biomaterials is discussed, including carbon-based nanomaterials, gold nanoparticles, graphite carbon nitride, transition metal dichalcogenides, and up-conversion nanoparticles that are used in the design of light-responsive medical devices. The importance of these light-responsive biomaterials is explored to design light-guided nanovehicle, modulate cellular behavior, as well as regulate extracellular microenvironments. Additionally, future perspectives on the clinical use of light-responsive biomaterials are highlighted.
Collapse
Affiliation(s)
- Hung Pang Lee
- Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Akhilesh K. Gaharwar
- Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
- Material Science and EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
- Center for Remote Health Technologies and SystemsTexas A&M UniversityCollege StationTX77843USA
| |
Collapse
|
17
|
P Yiu HH, Chari DM. How can nanoparticles help neural cell transplantation therapy? Nanomedicine (Lond) 2020; 15:2103-2106. [PMID: 32787617 DOI: 10.2217/nnm-2020-0279] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Humphrey H P Yiu
- Chemical Engineering, School of Engineering & Physical Sciences, Heriot-Watt University, Edinburgh, EH14 4AS, UK
| | - Divya M Chari
- Neural Tissue Engineering Keele group, School of Medicine, Keele University, Staffordshire, ST5 5BG, UK
| |
Collapse
|
18
|
Asgari V, Landarani-Isfahani A, Salehi H, Amirpour N, Hashemibeni B, Kazemi M, Bahramian H. Direct Conjugation of Retinoic Acid with Gold Nanoparticles to Improve Neural Differentiation of Human Adipose Stem Cells. J Mol Neurosci 2020; 70:1836-1850. [PMID: 32514739 DOI: 10.1007/s12031-020-01577-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 04/30/2020] [Indexed: 12/13/2022]
Abstract
Gold nanoparticles (AuNPs) have been proposed as useful medical carriers in the field of regenerative medicine. This study aimed to assess the direct conjugation ability of retinoic acid (RA) with AuNPs and to develop a strategy to differentiate the human adipose-derived stromal/stem cells (hADSCs) into neurons using AuNPs-RA. The physical properties of this nanocarrier were characterized using FT-IR, TEM, and FE-SEM. Moreover, the efficiency of RA conjugation on AuNPs was determined at 99% using UV-Vis spectroscopy. According to the MTT assay, an RA concentration of 66 μM caused a 50% inhibition of cell viability and AuNPs were not cytotoxic in concentrations below 5 μg/ml. Real-time PCR and immunocytochemistry proved that AuNPs-RA is able to increase the expression of neuronal marker genes and the number of neuronal protein (GFAP and MAP2)-positive cells, 14 days post-induction of hADSCs. Taken together, these results confirmed that the AuNPs-RA promote the neuronal differentiation of hADSCs.
Collapse
Affiliation(s)
- Vajihe Asgari
- Department of Anatomical Sciences, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Hossein Salehi
- Department of Anatomical Sciences, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Noushin Amirpour
- Department of Anatomical Sciences, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Batool Hashemibeni
- Department of Anatomical Sciences, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Kazemi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Bahramian
- Department of Anatomical Sciences, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
19
|
Yasodharababu M, Nair AK. A Multiscale Model to Predict Neuronal Cell Deformation with Varying Extracellular Matrix Stiffness and Topography. Cell Mol Bioeng 2020; 13:229-245. [PMID: 32426060 PMCID: PMC7225237 DOI: 10.1007/s12195-020-00615-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 04/11/2020] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Neuronal cells are sensitive to mechanical properties of extracellular matrix (ECM) such as stiffness and topography. Cells contract and exert a force on ECM to detect the microenvironment, which activates the signaling pathway to influence the cell functions such as differentiation, migration, and proliferation. There are numerous transmembrane proteins that transmit signals; however, integrin and neural cellular adhesion molecules (NCAM) play an important role in sensing the ECM mechanical properties. Mechanotransduction of cell-ECM is the key to understand the influence of ECM stiffness and topography; therefore, in this study, we develop a multiscale computational model to investigate these properties. METHODS This model couples the molecular behavior of integrin and NCAM to microscale interactions of neuronal cell and the ECM. We analyze the atomistic/molecular behavior of integrin and NCAM due to mechanical stimuli using steered molecular dynamics. The microscale properties of the neuronal cell and the ECM are simulated using non-linear finite element analysis by applying cell contractility. RESULTS We predict that by increasing the ECM stiffness, a neuronal cell exerts greater stress on the ECM. However, this stress reaches a saturation value for a threshold stiffness of ECM, and the saturation value is affected by the ECM thickness, topography, and clustering of integrin and NCAMs. Further, the ECM topography leads to asymmetric stress and deformation in the neuronal cell. Predicted stress distribution in neuronal cell and ECM are consistent with experimental results from the literature. CONCLUSION The multiscale computational model will guide in selecting the optimal ECM stiffness and topography range for in vitro studies.
Collapse
Affiliation(s)
- Mohan Yasodharababu
- Multiscale Materials Modeling Lab, Department of Mechanical Engineering, University of Arkansas, Fayetteville, AR USA
| | - Arun K. Nair
- Multiscale Materials Modeling Lab, Department of Mechanical Engineering, University of Arkansas, Fayetteville, AR USA
- Institute for Nanoscience and Engineering, University of Arkansas, 731 W. Dickson Street, Fayetteville, AR USA
| |
Collapse
|
20
|
Jiang J, Dhakal NP, Guo Y, Andre C, Thompson L, Skalli O, Peng C. Controlled Dynamics of Neural Tumor Cells by Templated Liquid Crystalline Polymer Networks. Adv Healthc Mater 2020; 9:e2000487. [PMID: 32378330 DOI: 10.1002/adhm.202000487] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/09/2020] [Indexed: 01/25/2023]
Abstract
The ability to control the alignment and organization of cell populations has great potential for tissue engineering and regenerative medicine. A variety of approaches such as nano/microtopographical patterning, mechanical loading, and nanocomposite synthesis have been developed to engineer scaffolds able to control cellular properties and behaviors. In this work, a patterned liquid crystal polymer network (LCN) film is synthesized by using a nematic liquid crystal template in which the molecular orientations are predesigned by photopatterning technique. Various configurations of polymer networks such as linear and circular patterns are created. When neural tumor cells are plated onto the templated LCN films, the cell alignment, migration, and proliferation are directed in both linear and curvilinear fashions following the pattern of the aligned polymer chains. A complex LCN pattern with zigzag geometry is also fabricated and found to be capable of controlling cell alignment and collective cellular organization. The demonstrated control of cell dynamics and organization by LCN films with various molecular alignments opens new opportunities to design scaffolds to control cultured cell organization in a manner resembling that found in tissues and to develop novel advanced materials for nerve repair, tissue engineering, and regenerative medicine applications.
Collapse
Affiliation(s)
- Jinghua Jiang
- Department of Physics and Materials ScienceThe University of Memphis Memphis TN 38152 USA
| | - Netra Prasad Dhakal
- Department of Physics and Materials ScienceThe University of Memphis Memphis TN 38152 USA
| | - Yubing Guo
- Advanced Materials and Liquid Crystal InstituteKent State University Kent OH 44242 USA
| | - Christian Andre
- Department of Physics and Materials ScienceThe University of Memphis Memphis TN 38152 USA
| | - Lauren Thompson
- Department of BiologyThe University of Memphis Memphis TN 38152 USA
| | - Omar Skalli
- Department of BiologyThe University of Memphis Memphis TN 38152 USA
| | - Chenhui Peng
- Department of Physics and Materials ScienceThe University of Memphis Memphis TN 38152 USA
| |
Collapse
|
21
|
Zhang S, Hang Y, Wu J, Tang Z, Li X, Zhang S, Wang L, Brash JL, Chen H. Dual Pathway for Promotion of Stem Cell Neural Differentiation Mediated by Gold Nanocomposites. ACS APPLIED MATERIALS & INTERFACES 2020; 12:22066-22073. [PMID: 32223207 DOI: 10.1021/acsami.9b22258] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The neural differentiation of embryonic stem cells (ESCs) is of great value in the treatment of neurodegenerative diseases. On the basis of the two related signaling pathways that direct the neural differentiation of ESCs, we used gold nanoparticles (GNP) as a means of combining chemical and physical cues to trigger the neurogenic differentiation of stem cells. Neural differentiation-related functional units (glyco and sulfonate units on glycosaminoglycans, GAG) were anchored on the GNP surface and were then transferred to the cell membrane surface via GNP-membrane interactions. The functional units were able to activate the GAG-related signaling pathway, in turn promoting differentiation and maturation of stem cells into neuronal lineages. In addition, using the photothermal effect of GNP, the differentiation-inducing factor retinoic acid (RA), could be actively delivered into cells via laser irradiation. The RA-related intracellular signaling pathway was thereby further triggered, resulting in strong promotion of neurogenesis with a 300-fold increase in mature neural marker expression. The gold nanocomposites developed in this work provide the basis for a new strategy directing ESCs differentiation into nerve cells with high efficiency and high purity by acting on two related signaling pathways.
Collapse
Affiliation(s)
- Sixuan Zhang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, P. R. China
| | - Yingjie Hang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, P. R. China
| | - Jingxian Wu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, P. R. China
| | - Zengchao Tang
- Jiangsu Biosurf Biotech Company, Ltd., Suzhou 215123, P. R. China
| | - Xin Li
- Suzhou Seemine-Nebula Biotech Company, Ltd., Suzhou 215123, P. R. China
| | - Shenghan Zhang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, P. R. China
| | - Lei Wang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, P. R. China
| | - John L Brash
- School of Biomedical Engineering and Department of Chemical Engineering, McMaster University, Hamilton, Ontario L8S4L7, Canada
| | - Hong Chen
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, P. R. China
| |
Collapse
|
22
|
Liang X, Li L, Tang J, Komiyama M, Ariga K. Dynamism of Supramolecular DNA/RNA Nanoarchitectonics: From Interlocked Structures to Molecular Machines. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2020. [DOI: 10.1246/bcsj.20200012] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Xingguo Liang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, P. R. China
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, P. R. China
| | - Lin Li
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, P. R. China
| | - Jiaxuan Tang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, P. R. China
| | - Makoto Komiyama
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, P. R. China
| | - Katsuhiko Ariga
- WPI-MANA, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
- Department of Advanced Materials Science, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8561, Japan
| |
Collapse
|
23
|
Cembran A, Bruggeman KF, Williams RJ, Parish CL, Nisbet DR. Biomimetic Materials and Their Utility in Modeling the 3-Dimensional Neural Environment. iScience 2020; 23:100788. [PMID: 31954980 PMCID: PMC6970178 DOI: 10.1016/j.isci.2019.100788] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 10/30/2019] [Accepted: 12/13/2019] [Indexed: 02/06/2023] Open
Abstract
The brain is a complex 3-dimensional structure, the organization of which provides a local environment that directly influences the survival, proliferation, differentiation, migration, and plasticity of neurons. To probe the effects of damage and disease on these cells, a synthetic environment is needed. Three-dimensional culturing of stem cells, neural progenitors, and neurons within fabricated biomaterials has demonstrated superior biomimetic properties over conventional 2-dimensional cultureware, offering direct recapitulation of both cell-cell and cell-extracellular matrix interactions. Within this review we address the benefits of deploying biomaterials as advanced cell culture tools capable of influencing neuronal fate and as in vitro models of the native in vivo microenvironment. We highlight recent and promising biomaterials approaches toward understanding neural network and their function relevant to neurodevelopment and provide our perspective on how these materials can be engineered and programmed to study both the healthy and diseased nervous system.
Collapse
Affiliation(s)
- Arianna Cembran
- Laboratory of Advanced Biomaterials, Research School of Electrical, Energy and Materials Engineering, The Australian National University, Canberra, ACT 2600, Australia
| | - Kiara F Bruggeman
- Laboratory of Advanced Biomaterials, Research School of Electrical, Energy and Materials Engineering, The Australian National University, Canberra, ACT 2600, Australia
| | | | - Clare L Parish
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Melbourne, VIC 3010, Australia.
| | - David R Nisbet
- Laboratory of Advanced Biomaterials, Research School of Electrical, Energy and Materials Engineering, The Australian National University, Canberra, ACT 2600, Australia.
| |
Collapse
|
24
|
Maysinger D, Ji J. Nanostructured Modulators of Neuroglia. Curr Pharm Des 2019; 25:3905-3916. [PMID: 31512994 DOI: 10.2174/1381612825666190912163339] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/08/2019] [Indexed: 01/08/2023]
Abstract
Biological and synthetic nanostructures can influence both glia and neurons in the central nervous system. Neurons represent only a small proportion (about 10%) of cells in the brain, whereas glial cells are the most abundant cell type. Non-targeted nanomedicines are mainly internalized by glia, in particular microglia, and to a lesser extent by astrocytes. Internalized nanomedicines by glia indirectly modify the functional status of neurons. The mechanisms of biochemical, morphological and functional changes of neural cells exposed to nanomedicines are still not well-understood. This minireview provides a cross-section of morphological and biochemical changes in glial cells and neurons exposed to different classes of hard and soft nanostructures.
Collapse
Affiliation(s)
- Dusica Maysinger
- Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, Quebec H3AOG4, Canada
| | - Jeff Ji
- Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, Quebec H3AOG4, Canada
| |
Collapse
|
25
|
High-Dose Neural Stem/Progenitor Cell Transplantation Increases Engraftment and Neuronal Distribution and Promotes Functional Recovery in Rats after Acutely Severe Spinal Cord Injury. Stem Cells Int 2019; 2019:9807978. [PMID: 31565061 PMCID: PMC6745168 DOI: 10.1155/2019/9807978] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 11/04/2018] [Indexed: 01/12/2023] Open
Abstract
Severe spinal cord injury (SCI) leads to permanent, complete paraplegia and places considerable mental and economic burdens on patients, compared with mild to moderate SCI. However, the dose-related effects of the neural stem/precursor cell (NSPC) transplantation on the injury microenvironment, NSPC survival, axonal growth, neuronal distribution, the composition of neurons, oligodendrocytes, and astrocytes in the lesion area and functional recovery have not yet been quantitatively evaluated in the context of severe SCI. In our study, we acutely transplanted 2.5 × 104 or 1.5 × 105 NSPCs/μl into the site of transection SCI. We found that high-dose NSPC transplantation exerted immunomodulatory and neuroprotective effects in the acute phase of severe SCI. In addition, one week later, a remarkable positive relationship was observed between the transplantation dose and the number of surviving NSPCs in severe SCI. At 8 weeks postgrafting, subjects that received the higher cell dose exhibited abundant nerve regeneration, extensive neuronal distribution, increased proportions of neurons and oligodendrocytes, and nascent functional neural network formation in the lesion area. Notably, a significant functional recovery was also observed. Our data suggest that it is important to consider potential dose-related effects on donor cell survival, neuronal distribution, and locomotor recovery in the development of preclinical NSPC transplantation therapy for severe SCI.
Collapse
|
26
|
Sun H, Yu D, Guan Y, Du Z, Ren J, Qu X. Wireless near-infrared electrical stimulation of neurite outgrowth. Chem Commun (Camb) 2019; 55:9833-9836. [PMID: 31363722 DOI: 10.1039/c9cc03537k] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Herein, through using electropolymerized pyrrole (PPy) to coat near-infrared upconversion nanoparticles (UCNPs) on an indium tin oxide (ITO) electrode, the as-prepared PPy/UCNPs photoelectrode could generate an interfacial electric field, release rare earth ions and induce reactive oxygen species (ROS) in PC12 cells under NIR irradiation, which could realize wireless neurite development and outgrowth.
Collapse
Affiliation(s)
- Hanjun Sun
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China.
| | | | | | | | | | | |
Collapse
|
27
|
Che Marzuki NH, Wahab RA, Abdul Hamid M. An overview of nanoemulsion: concepts of development and cosmeceutical applications. BIOTECHNOL BIOTEC EQ 2019. [DOI: 10.1080/13102818.2019.1620124] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Affiliation(s)
- Nur Haziqah Che Marzuki
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia (UTM), Johor Bahru, Malaysia
- Enzyme Technology and Green Synthesis Group, Faculty of Science, Universiti Teknologi Malaysia (UTM), Johor Bahru, Malaysia
| | - Roswanira Abdul Wahab
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia (UTM), Johor Bahru, Malaysia
- Enzyme Technology and Green Synthesis Group, Faculty of Science, Universiti Teknologi Malaysia (UTM), Johor Bahru, Malaysia
| | - Mariani Abdul Hamid
- Department of Engineering, School of Chemical & Energy Engineering, Institute of Bioproduct Development, Universiti Teknologi Malaysia (UTM), Johor Bahru, Malaysia
| |
Collapse
|
28
|
Sun G, Yang S, Cai H, Shu Y, Han Q, Wang B, Li Z, Zhou L, Gao Q, Yin Z. Molybdenum disulfide nanoflowers mediated anti-inflammation macrophage modulation for spinal cord injury treatment. J Colloid Interface Sci 2019; 549:50-62. [PMID: 31015056 DOI: 10.1016/j.jcis.2019.04.047] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 04/15/2019] [Accepted: 04/15/2019] [Indexed: 12/11/2022]
Abstract
Spinal cord injury (SCI) can cause locomotor dysfunctions and sensory deficits. Evidence shows that functional nanodrugs can regulate macrophage polarization and promote anti-inflammatory cytokine expression, which is feasible in SCI immunotherapeutic treatments. Molybdenum disulfide (MoS2) nanomaterials have garnered great attention as potential carriers for therapeutic payload. Herein, we synthesize MoS2@PEG (MoS2 = molybdenum disulfide, PEG = poly (ethylene glycol)) nanoflowers as an effective carrier for loading etanercept (ET) to treat SCI. We characterize drug loading and release properties of MoS2@PEG in vitro and demonstrate that ET-loading MoS2@PEG obviously inhibits the expression of M1-related pro-inflammatory markers (TNF-α, CD86 and iNOS), while promoting M2-related anti-inflammatory markers (Agr1, CD206 and IL-10) levels. In vivo, the mouse model of SCI shows that long-circulating ET-MoS2@PEG nanodrugs can effectively extravasate into the injured spinal cord up to 96 h after SCI, and promote macrophages towards M2 type polarization. As a result, the ET-loading MoS2@PEG administration in mice can protect survival motor neurons, thus, reducing injured areas at central lesion sites, and significantly improving locomotor recovery. This study demonstrates the anti-inflammatory and neuroprotective activities of ET-MoS2@PEG and promising utility of MoS2 nanomaterial-mediated drug delivery.
Collapse
Affiliation(s)
- Guodong Sun
- The First Affiliated Hospital, Biomedical Translational Research Institute, Jinan University, Guangzhou 510632, PR China
| | - Shuxian Yang
- The First Affiliated Hospital, Biomedical Translational Research Institute, Jinan University, Guangzhou 510632, PR China
| | - Huaihong Cai
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, PR China
| | - Yijin Shu
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, PR China
| | - Qi Han
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Baocheng Wang
- The First Affiliated Hospital, Biomedical Translational Research Institute, Jinan University, Guangzhou 510632, PR China
| | - Zhizhong Li
- The First Affiliated Hospital, Jinan University, Guangzhou 510632, PR China
| | - Libing Zhou
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou 510632, PR China
| | - Qingsheng Gao
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, PR China.
| | - Zhinan Yin
- The First Affiliated Hospital, Biomedical Translational Research Institute, Jinan University, Guangzhou 510632, PR China.
| |
Collapse
|
29
|
Henrich-Noack P, Nikitovic D, Neagu M, Docea AO, Engin AB, Gelperina S, Shtilman M, Mitsias P, Tzanakakis G, Gozes I, Tsatsakis A. The blood–brain barrier and beyond: Nano-based neuropharmacology and the role of extracellular matrix. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 17:359-379. [DOI: 10.1016/j.nano.2019.01.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/11/2019] [Accepted: 01/28/2019] [Indexed: 12/13/2022]
|
30
|
Mohammadniaei M, Yoon J, Choi HK, Placide V, Bharate BG, Lee T, Choi JW. Multifunctional Nanobiohybrid Material Composed of Ag@Bi 2Se 3/RNA Three-Way Junction/miRNA/Retinoic Acid for Neuroblastoma Differentiation. ACS APPLIED MATERIALS & INTERFACES 2019; 11:8779-8788. [PMID: 30714374 DOI: 10.1021/acsami.8b16925] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Nanoparticle-based cell differentiation therapy has attracted increasing research interest as it is a promising substitute for conventional cancer treatment methods. Here, the topological insulator bismuth selenide nanoparticle (Bi2Se3 NP) was core-shelled with silver (Ag@Bi2Se3) to represent remarkable biocompatibility and plasmonic features (ca. 2.3 times higher than those of Ag nanoparticle). Moreover, a newly developed RNA three-way junction (3WJ) structure was designed for the quad-functionalization of any type of nanoparticle and surface. One leg of the 3WJ was attached to the Ag@Bi2Se3, and the other leg harbored a cell-penetrating RNA and a florescence tag. The third leg was designed to inhibit micro-RNA-17 (miR-17) and to further release retinoic acid (RA). A new drug delivery mechanism was developed for the slow release of RA inside the cytosol based on the prerequisite inhibition of miR-17 using a strand displacement strategy. In this paper, we report a simple methodology for resolving the hydrophobicity challenges of RA by its conjugation with a RNA strand (RA/R) through a stimulus-responsive cross-linker. The developed nanobiohybrid material could fully differentiate SH-SY5Y cancer cells into neurons and stop their growth in 6 days without requiring sequential treatments which has not been reported yet. Using a surface-enhanced Raman spectroscopy technique, the RA delivery and the cell differentiation process were monitored nondestructively in real time. The fabricated nanobiohybrid material could open the new horizons in the fabrication of different diagnostic/therapeutic agents.
Collapse
Affiliation(s)
- Mohsen Mohammadniaei
- Department of Chemical and Biomolecular Engineering , Sogang University , 35 Baekbeom-ro (Sinsu-dong) , Mapo-gu, Seoul 121-742 , Republic of Korea
| | - Jinho Yoon
- Department of Chemical and Biomolecular Engineering , Sogang University , 35 Baekbeom-ro (Sinsu-dong) , Mapo-gu, Seoul 121-742 , Republic of Korea
| | - Hye Kyu Choi
- Department of Chemical and Biomolecular Engineering , Sogang University , 35 Baekbeom-ro (Sinsu-dong) , Mapo-gu, Seoul 121-742 , Republic of Korea
| | - Virginie Placide
- Department of Chemical and Biomolecular Engineering , Sogang University , 35 Baekbeom-ro (Sinsu-dong) , Mapo-gu, Seoul 121-742 , Republic of Korea
| | - Bapurao Gangaram Bharate
- Department of Chemical and Biomolecular Engineering , Sogang University , 35 Baekbeom-ro (Sinsu-dong) , Mapo-gu, Seoul 121-742 , Republic of Korea
| | - Taek Lee
- Department of Chemical Engineering , Kwangwoon University , 20 Kwangwoon-ro , Nowon-gu, Seoul 01897 , Republic of Korea
| | - Jeong-Woo Choi
- Department of Chemical and Biomolecular Engineering , Sogang University , 35 Baekbeom-ro (Sinsu-dong) , Mapo-gu, Seoul 121-742 , Republic of Korea
| |
Collapse
|
31
|
Wu Y, Peng Y, Bohra H, Zou J, Ranjan VD, Zhang Y, Zhang Q, Wang M. Photoconductive Micro/Nanoscale Interfaces of a Semiconducting Polymer for Wireless Stimulation of Neuron-Like Cells. ACS APPLIED MATERIALS & INTERFACES 2019; 11:4833-4841. [PMID: 30624894 DOI: 10.1021/acsami.8b19631] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
We report multiscale structured fibers and patterned films based on a semiconducting polymer, poly(3-hexylthiophene) (P3HT), as photoconductive biointerfaces to promote neuronal stimulation upon light irradiation. The micro/nanoscale structures of P3HT used for neuronal interfacing and stimulation include nanofibers with an average diameter of 100 nm, microfibers with an average diameter of about 1 μm, and lithographically patterned stripes with width of 3, 25, and 50 μm, respectively. The photoconductive effect of P3HT upon light irradiation provides electrical stimulation for neuronal differentiation and directed growth. Our results demonstrate that neurons on P3HT nanofibers showed a significantly higher total number of branches, while neurons grown on P3HT microfibers had longer and thinner neurites. Such a combination strategy of topographical and photoconductive stimulation can be applied to further enhance neuronal differentiation and directed growth. These photoconductive polymeric micro/nanostructures demonstrated their great potential for neural engineering and development of novel neural regenerative devices.
Collapse
Affiliation(s)
- Yingjie Wu
- School of Chemical and Biomedical Engineering , Nanyang Technological University , 62 Nanyang Drive , Singapore 637459 , Singapore
| | - Yanfen Peng
- School of Chemical and Biomedical Engineering , Nanyang Technological University , 62 Nanyang Drive , Singapore 637459 , Singapore
| | - Hassan Bohra
- School of Chemical and Biomedical Engineering , Nanyang Technological University , 62 Nanyang Drive , Singapore 637459 , Singapore
| | - Jianping Zou
- School of Electrical and Electronic Engineering , Nanyang Technological University , 50 Nanyang Avenue , Singapore 639798 , Singapore
| | - Vivek Damodar Ranjan
- School of Mechanical & Aerospace Engineering , Nanyang Technological University , 50 Nanyang Avenue , Singapore 639798 , Singapore
| | - Yilei Zhang
- School of Mechanical & Aerospace Engineering , Nanyang Technological University , 50 Nanyang Avenue , Singapore 639798 , Singapore
| | - Qing Zhang
- School of Electrical and Electronic Engineering , Nanyang Technological University , 50 Nanyang Avenue , Singapore 639798 , Singapore
| | - Mingfeng Wang
- School of Chemical and Biomedical Engineering , Nanyang Technological University , 62 Nanyang Drive , Singapore 637459 , Singapore
| |
Collapse
|
32
|
Wang Z, Hu M, Ai X, Zhang Z, Xing B. Near-Infrared Manipulation of Membrane Ion Channels via Upconversion Optogenetics. ADVANCED BIOSYSTEMS 2019; 3:e1800233. [PMID: 32627341 DOI: 10.1002/adbi.201800233] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 09/27/2018] [Indexed: 12/21/2022]
Abstract
Membrane ion channels are ultimately responsible for the propagation and integration of electrical signals in the nervous, muscular, and other systems. Their activation or malfunctioning plays a significant role in physiological and pathophysiological processes. Using optogenetics to dynamically and spatiotemporally control ion channels has recently attracted considerable attention. However, most of the established optogenetic tools (e.g., channelrhodopsins, ChRs) for optical manipulations, are mainly stimulated by UV or visible light, which raises the concerns of potential photodamage, limited tissue penetration, and high-invasive implantation of optical fiber devices. Near-infrared (NIR) upconversion nanoparticle (UCNP)-mediated optogenetic systems provide great opportunities for overcoming the problems encountered in the manipulation of ion channels in deep tissues. Hence, this review focuses on the recent advances in NIR regulation of membrane ion channels via upconversion optogenetics in biomedical research. The engineering and applications of upconversion optogenetic systems by the incorporation multiple emissive UCNPs into various light-gated ChRs/ligands are first elaborated, followed by a detailed discussion of the technical improvements for more precise and efficient control of membrane channels. Finally, the future perspectives for refining and advancing NIR-mediated upconversion optogenetics into in vivo even in clinical applications are proposed.
Collapse
Affiliation(s)
- Zhimin Wang
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University, Singapore, 637371, Singapore
| | - Ming Hu
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University, Singapore, 637371, Singapore
| | - Xiangzhao Ai
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University, Singapore, 637371, Singapore
| | - Zhijun Zhang
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University, Singapore, 637371, Singapore
| | - Bengang Xing
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University, Singapore, 637371, Singapore
| |
Collapse
|
33
|
Zhang Z, Klausen LH, Chen M, Dong M. Electroactive Scaffolds for Neurogenesis and Myogenesis: Graphene-Based Nanomaterials. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1801983. [PMID: 30264534 DOI: 10.1002/smll.201801983] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 07/28/2018] [Indexed: 05/24/2023]
Abstract
One of the major issues in tissue engineering is constructing a functional scaffold to support cell growth and also provide proper synergistic guidance cues. Graphene-based nanomaterials have emerged as biocompatible and electroactive scaffolds for neurogenesis and myogenesis, due to their excellent tunable chemical, physical, and mechanical properties. This review first assesses the recent investigations focusing on the fabrication and applications of graphene-based nanomaterials for neurogenesis and myogenesis, in the form of either 2D films, 3D scaffolds, or composite architectures. Besides, because of their outstanding electrical properties, graphene family materials are particularly suitable for designing electroactive scaffolds that could provide proper electrical stimulation (i.e., electrical or photo stimuli) to promote the regeneration of excitable neurons and muscle cells. Therefore, the effects and mechanism of electrical and/or photo stimulations on neurogenesis and myogenesis are followed. Furthermore, studies on their biocompatibilities and toxicities especially to neural and muscle cells are evaluated. Finally, the future challenges and perspectives in facilitating the development of clinical translation of graphene-family nanomaterials in treating neurodegenerative and muscle diseases are discussed.
Collapse
Affiliation(s)
- Zhongyang Zhang
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, DK-8000, Aarhus C, Denmark
| | | | - Menglin Chen
- Department of Engineering, Aarhus University, DK-8000, Aarhus C, Denmark
| | - Mingdong Dong
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, DK-8000, Aarhus C, Denmark
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
34
|
Pardo-Figuerez M, Martin NRW, Player DJ, Roach P, Christie SDR, Capel AJ, Lewis MP. Controlled Arrangement of Neuronal Cells on Surfaces Functionalized with Micropatterned Polymer Brushes. ACS OMEGA 2018; 3:12383-12391. [PMID: 30411006 PMCID: PMC6217525 DOI: 10.1021/acsomega.8b01698] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 09/17/2018] [Indexed: 05/03/2023]
Abstract
Conventional in vitro cultures are useful to represent simplistic neuronal behavior; however, the lack of organization results in random neurite spreading. To overcome this problem, control over the directionality of SH-SY5Y cells was attained, utilizing photolithography to pattern the cell-repulsive anionic brush poly(potassium 3-sulfopropyl methacrylate) (PKSPMA) into tracks of 20, 40, 80, and 100 μm width. These data validate the use of PKSPMA brush coatings for a long-term culture of the SH-SY5Y cells, as well as providing a methodology by which the precise deposition of PKSPMA can be utilized to achieve a targeted control over the SH-SY5Y cells. Specifically, the PKSPMA brush patterns prevented cell attachment, allowing the SH-SY5Y cells to grow only on noncoated glass (gaps of 20, 50, 75, and 100 μm width) at different cell densities (5000, 10 000, and 15 000 cells/cm2). This research demonstrates the importance of achieving cell directionality in vitro, while these simplistic models could provide new platforms to study complex neuron-neuron interactions.
Collapse
Affiliation(s)
- Maria Pardo-Figuerez
- National
Centre for Sport and Exercise Medicine (NCSEM), School of
Sport, Exercise and Health Sciences, and Department of Chemistry, School
of Science, Loughborough University, Loughborough LE11 3TU, U.K.
| | - Neil R. W. Martin
- National
Centre for Sport and Exercise Medicine (NCSEM), School of
Sport, Exercise and Health Sciences, and Department of Chemistry, School
of Science, Loughborough University, Loughborough LE11 3TU, U.K.
| | - Darren J. Player
- National
Centre for Sport and Exercise Medicine (NCSEM), School of
Sport, Exercise and Health Sciences, and Department of Chemistry, School
of Science, Loughborough University, Loughborough LE11 3TU, U.K.
- Institute
of Orthopaedics and Musculoskeletal Science, University College London, Stanmore HA7 4LP, U.K.
| | - Paul Roach
- National
Centre for Sport and Exercise Medicine (NCSEM), School of
Sport, Exercise and Health Sciences, and Department of Chemistry, School
of Science, Loughborough University, Loughborough LE11 3TU, U.K.
| | - Steven D. R. Christie
- National
Centre for Sport and Exercise Medicine (NCSEM), School of
Sport, Exercise and Health Sciences, and Department of Chemistry, School
of Science, Loughborough University, Loughborough LE11 3TU, U.K.
| | - Andrew J. Capel
- National
Centre for Sport and Exercise Medicine (NCSEM), School of
Sport, Exercise and Health Sciences, and Department of Chemistry, School
of Science, Loughborough University, Loughborough LE11 3TU, U.K.
| | - Mark P. Lewis
- National
Centre for Sport and Exercise Medicine (NCSEM), School of
Sport, Exercise and Health Sciences, and Department of Chemistry, School
of Science, Loughborough University, Loughborough LE11 3TU, U.K.
| |
Collapse
|
35
|
Jiang J, Wang Y, Liu B, Chen X, Zhang S. Challenges and research progress of the use of mesenchymal stem cells in the treatment of ischemic stroke. Brain Dev 2018; 40:612-626. [PMID: 29661589 DOI: 10.1016/j.braindev.2018.03.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/28/2018] [Accepted: 03/29/2018] [Indexed: 02/06/2023]
Abstract
Cerebral Ischemic Stroke (CIS) has become a hot issue in medical research because of the diversity of risk factors and the uncertainty of prognosis. In the field of regenerative medicine, mesenchymal stem cells (MSCs) have an increasingly prominent position due to their advantages of multiple differentiation, low immunogenicity and wide application. In the basic and clinical research of CIS, there are still some problems to be solved in the treatment of CIS. This paper will discuss the progresses and some obstacles of current MSCs for the treatment of CIS.
Collapse
Affiliation(s)
- Jipeng Jiang
- Institution of Brain Trauma and Neurology Disease of Affiliated Hospital of Logistics University of People's Armed Police Forces, Chenglin Road No. 220, Tianjin 300162, China.
| | - Yuting Wang
- Tianjin Medical University, Qixiangtai Road No. 22, Tianjin 300070, China
| | - Baohu Liu
- Tianjin University of Traditional Chinese Medicine, Yuquan Road No. 88, Tianjin 300193, China
| | - Xuyi Chen
- Institution of Brain Trauma and Neurology Disease of Affiliated Hospital of Logistics University of People's Armed Police Forces, Chenglin Road No. 220, Tianjin 300162, China
| | - Sai Zhang
- Institution of Brain Trauma and Neurology Disease of Affiliated Hospital of Logistics University of People's Armed Police Forces, Chenglin Road No. 220, Tianjin 300162, China.
| |
Collapse
|
36
|
Komiyama M, Mori T, Ariga K. Molecular Imprinting: Materials Nanoarchitectonics with Molecular Information. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2018. [DOI: 10.1246/bcsj.20180084] [Citation(s) in RCA: 161] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Makoto Komiyama
- WPI-MANA, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
- Life Science Center of Tsukuba Advanced Research Alliance, University of Tsukuba, 1-1-1 Ten-noudai, Tsukuba, Ibaraki 305-8577, Japan
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, P. R. China
| | - Taizo Mori
- WPI-MANA, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Katsuhiko Ariga
- WPI-MANA, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
- Department of Advanced Materials Science, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8561, Japan
| |
Collapse
|
37
|
Li B, Agarwal V, Ho D, Vede JP, Iyer KS. Systematic assessment of surface functionality on nanoscale patterns for topographic contact guidance of cells. NEW J CHEM 2018. [DOI: 10.1039/c7nj04914e] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The ability of surface topography to influence cellular response has been widely accepted, leading the way towards the development of potential neural prosthetics.
Collapse
Affiliation(s)
- Binbin Li
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing
- Wuhan University of Technology
- Wuhan 430070
- P. R. China
- Biomedical Materials and Engineering Research Center of Hubei Province
| | - Vipul Agarwal
- School of Molecular Sciences
- The University of Western Australia
- Crawley WA 6009
- Australia
| | - Dominic Ho
- School of Molecular Sciences
- The University of Western Australia
- Crawley WA 6009
- Australia
| | | | - K. Swaminathan Iyer
- School of Molecular Sciences
- The University of Western Australia
- Crawley WA 6009
- Australia
| |
Collapse
|
38
|
Lefèvre D, Louvegny J, Naudin M, Ferain E, Dupont-Gillain C, Demoustier-Champagne S. Biofunctionalized and self-supported polypyrrole frameworks as nanostructured ECM-like biointerfaces. RSC Adv 2018; 8:22932-22943. [PMID: 35540120 PMCID: PMC9081635 DOI: 10.1039/c8ra00325d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 06/13/2018] [Indexed: 11/21/2022] Open
Abstract
Hybrid nanobiointerfaces were designed as an original contribution to the challenge of synthesizing nanostructured biomaterials integrating a set of cell fate-determining cues, originally provided to cells by the extracellular matrix (ECM). The produced biointerfaces consist of a stiff framework of intersected polypyrrole (PPy) nanotubes supporting a soft multilayer composed of ECM-derived biomacromolecules: collagen (Col) and hyaluronic acid (HA). PPy frameworks with highly tunable characteristics were synthesized through chemical oxidative polymerization of pyrrole monomers, templated within track-etched polycarbonate (PC) membranes featuring a network of intersected nanopores. PPy interfaces with a porosity of 80%, composed of nanotubes with an average diameter ranging from 40 to 300 nm, intersecting at an angle of 90°, were shown to be self-supported. These rigid PPy nanostructured interfaces were functionalized with a self-assembling (HA/Col) multilayer deposited via a layer-by-layer process. Biofunctionalized and unmodified PPy frameworks were both shown to promote sustained cell adhesion, therefore demonstrating the cytocompatibility of the engineered matrices. Such nanobiointerfaces, combining a mechanically-stable framework of tunable dimensions with a soft biopolymeric multilayer of highly versatile nature, pave the way towards cell-instructive biomaterials able to gather a wide range of cues guiding cell behavior. The developed self-supported structures could be used as a coating or as membranes bridging different tissues. A versatile template-based approach allows for the synthesis of nanostructured biointerfaces, made of core–shell nanotubes, combining bioactivity and mechanical stability.![]()
Collapse
Affiliation(s)
- Damien Lefèvre
- Institute of Condensed Matter and Nanosciences (Bio & Soft Matter)
- Louvain-la-Neuve
- Belgium
| | - Juliette Louvegny
- Institute of Condensed Matter and Nanosciences (Bio & Soft Matter)
- Louvain-la-Neuve
- Belgium
| | - Mathieu Naudin
- Institute of Condensed Matter and Nanosciences (Bio & Soft Matter)
- Louvain-la-Neuve
- Belgium
| | - Etienne Ferain
- Institute of Condensed Matter and Nanosciences (Bio & Soft Matter)
- Louvain-la-Neuve
- Belgium
- It4ip S.A
- Louvain-la-Neuve
| | | | | |
Collapse
|
39
|
Aydin T, Gurcan C, Taheri H, Yilmazer A. Graphene Based Materials in Neural Tissue Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1107:129-142. [PMID: 29882208 DOI: 10.1007/5584_2018_221] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Due to its extraordinary features such as large surface area, high electrical conductivity, chemical stability and mechanical properties, graphene attracts great interest in various fields of biomedical sciences including biosensors, cancer therapy, diagnosis and regenerative medicine. The use of graphene-based materials has been of great interest for the design of scaffolds that can promote neural tissue regeneration. Recent studies published over the last few years clearly show that graphene and graphene based materials promote adhesion, proliferation and differentiation of various cells including embryonic stem cells (ESC), neural stem cells (NSC), mesenchymal stem cells (MSC) and induced pluripotent stem cells (iPSC). Therefore graphene based materials are one of the promising nanoplatforms in regenerative medicine for neural tissue injury. With its unique topographic and chemical properties, graphene is used as a scaffold that could provide a bridge between regenerating nerves. More importantly, as a conductive substrate, graphene allows the continuation of electrical conduction between damaged nerve ends. The integration of supportive cells such as glial, neural precursor or stem cells in such a scaffold shows higher regeneration when compared to currently used neural autografts and nerve conduits. This review discusses the details of such studies involving graphene based materials with a special interest on neural stem cells, mesenchymal stem cells or pluripotent stem cells.
Collapse
Affiliation(s)
- Tugce Aydin
- Biotechnology Institute, Ankara University, Tandogan/Ankara, Turkey.,Engineering Faculty, Biomedical Engineering Department, Ankara University, Tandogan/Ankara, Turkey
| | - Cansu Gurcan
- Biotechnology Institute, Ankara University, Tandogan/Ankara, Turkey.,Engineering Faculty, Biomedical Engineering Department, Ankara University, Tandogan/Ankara, Turkey
| | - Hadiseh Taheri
- Engineering Faculty, Biomedical Engineering Department, Ankara University, Tandogan/Ankara, Turkey
| | - Açelya Yilmazer
- Engineering Faculty, Biomedical Engineering Department, Ankara University, Tandogan/Ankara, Turkey. .,Stem Cell Institute, Ankara University, Balgat/Ankara, Turkey.
| |
Collapse
|
40
|
Kumar A, Tan A, Wong J, Spagnoli JC, Lam J, Blevins BD, G N, Thorne L, Ashkan K, Xie J, Liu H. Nanotechnology for Neuroscience: Promising Approaches for Diagnostics, Therapeutics and Brain Activity Mapping. ADVANCED FUNCTIONAL MATERIALS 2017; 27:1700489. [PMID: 30853878 PMCID: PMC6404766 DOI: 10.1002/adfm.201700489] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Unlocking the secrets of the brain is a task fraught with complexity and challenge - not least due to the intricacy of the circuits involved. With advancements in the scale and precision of scientific technologies, we are increasingly equipped to explore how these components interact to produce a vast range of outputs that constitute function and disease. Here, an insight is offered into key areas in which the marriage of neuroscience and nanotechnology has revolutionized the industry. The evolution of ever more sophisticated nanomaterials culminates in network-operant functionalized agents. In turn, these materials contribute to novel diagnostic and therapeutic strategies, including drug delivery, neuroprotection, neural regeneration, neuroimaging and neurosurgery. Further, the entrance of nanotechnology into future research arenas including optogenetics, molecular/ion sensing and monitoring, and piezoelectric effects is discussed. Finally, considerations in nanoneurotoxicity, the main barrier to clinical translation, are reviewed, and direction for future perspectives is provided.
Collapse
Affiliation(s)
- Anil Kumar
- State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, China
| | - Aaron Tan
- UCL Medical School, University College London (UCL), London, United Kingdom
| | - Joanna Wong
- Imperial College School of Medicine, Imperial College London,London, United Kingdom
| | - Jonathan Clayton Spagnoli
- Department of Chemistry, Bio-Imaging Research Center, University of Georgia, Athens, Georgia 30602, United States
| | - James Lam
- UCL Medical School, University College London (UCL), London, United Kingdom
| | - Brianna Diane Blevins
- Department of Chemistry, Bio-Imaging Research Center, University of Georgia, Athens, Georgia 30602, United States
| | - Natasha G
- UCL Medical School, University College London (UCL), London, United Kingdom
| | - Lewis Thorne
- Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, Queen Square, London, United Kingdom
| | - Keyoumars Ashkan
- Department of Neurosurgery, King's College Hospital NHS Foundation Trust, King's College London, London, United Kingdom
| | - Jin Xie
- Department of Chemistry, Bio-Imaging Research Center, University of Georgia, Athens, Georgia 30602, United States
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, China
| |
Collapse
|
41
|
Majumdar R, Tantayanon S, Bag BG. Synthesis of palladium nanoparticles with leaf extract of Chrysophyllum cainito (Star apple) and their applications as efficient catalyst for C–C coupling and reduction reactions. INTERNATIONAL NANO LETTERS 2017. [DOI: 10.1007/s40089-017-0220-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
42
|
Komiyama M, Yoshimoto K, Sisido M, Ariga K. Chemistry Can Make Strict and Fuzzy Controls for Bio-Systems: DNA Nanoarchitectonics and Cell-Macromolecular Nanoarchitectonics. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2017. [DOI: 10.1246/bcsj.20170156] [Citation(s) in RCA: 238] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Makoto Komiyama
- World Premier International (WPI) Research Centre for Materials Nanoarchitectonics (MANA), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki 305-0044
- Life Science Center of Tsukuba Advanced Research Alliance, University of Tsukuba, 1-1-1 Ten-noudai, Tsukuba, Ibaraki 305-8577
| | - Keitaro Yoshimoto
- Department of Life Sciences, Graduate School of Arts and Science, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902
| | - Masahiko Sisido
- Professor Emeritus, Research Core for Interdisciplinary Sciences, Okayama University, 3-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530
| | - Katsuhiko Ariga
- World Premier International (WPI) Research Centre for Materials Nanoarchitectonics (MANA), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki 305-0044
- Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba 277-0827
| |
Collapse
|
43
|
Kumar R, Urtizberea A, Ghosh S, Bog U, Rainer Q, Lenhert S, Fuchs H, Hirtz M. Polymer Pen Lithography with Lipids for Large-Area Gradient Patterns. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2017. [PMID: 28650173 DOI: 10.1021/acs.langmuir.7b01368] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Gradient patterns comprising bioactive compounds over comparably (in regard to a cell size) large areas are key for many applications in the biomedical sector, in particular, for cell screening assays, guidance, and migration experiments. Polymer pen lithography (PPL) as an inherent highly parallel and large area technique has a great potential to serve in the fabrication of such patterns. We present strategies for the printing of functional phospholipid patterns via PPL that provide tunable feature size and feature density gradients over surface areas of several square millimeters. By controlling the printing parameters, two transfer modes can be achieved. Each of these modes leads to different feature morphologies. By increasing the force applied to the elastomeric pens, which increases the tip-surface contact area and boosts the ink delivery rate, a switch between a dip-pen nanolithography (DPN) and a microcontact printing (μCP) transfer mode can be induced. A careful inking procedure ensuring a homogeneous and not-too-high ink-load on the PPL stamp ensures a membrane-spreading dominated transfer mode, which, used in combination with smooth and hydrophilic substrates, generates features with constant height, independently of the applied force of the pens. Ultimately, this allows us to obtain a gradient of feature sizes over a mm2 substrate, all having the same height on the order of that of a biological cellular membrane. These strategies allow the construction of membrane structures by direct transfer of the lipid mixture to the substrate, without requiring previous substrate functionalization, in contrast to other molecular inks, where structure is directly determined by the printing process itself. The patterns are demonstrated to be viable for subsequent protein binding, therefore adding to a flexible feature library when gradients of protein presentation are desired.
Collapse
Affiliation(s)
- Ravi Kumar
- Institute of Nanotechnology (INT) and Karlsruhe Nano Micro Facility (KNMF), Karlsruhe Institute of Technology (KIT) , 76131 Karlsruhe, Germany
- Physical Institute and Center for Nanotechnology (CeNTech), University of Münster , 48149 Münster, Germany
| | - Ainhoa Urtizberea
- Institute of Nanotechnology (INT) and Karlsruhe Nano Micro Facility (KNMF), Karlsruhe Institute of Technology (KIT) , 76131 Karlsruhe, Germany
| | - Souvik Ghosh
- Institute of Nanotechnology (INT) and Karlsruhe Nano Micro Facility (KNMF), Karlsruhe Institute of Technology (KIT) , 76131 Karlsruhe, Germany
- Sardar Vallabhbhai National Institute of Technology (SVNIT) , Surat, Gujarat 395007, India
| | - Uwe Bog
- Institute of Nanotechnology (INT) and Karlsruhe Nano Micro Facility (KNMF), Karlsruhe Institute of Technology (KIT) , 76131 Karlsruhe, Germany
| | - Quinn Rainer
- Florida State Univ , Dept Biol Sci and Integrat NanoSci Inst, Tallahassee, Florida 32306 United States
| | - Steven Lenhert
- Florida State Univ , Dept Biol Sci and Integrat NanoSci Inst, Tallahassee, Florida 32306 United States
| | - Harald Fuchs
- Institute of Nanotechnology (INT) and Karlsruhe Nano Micro Facility (KNMF), Karlsruhe Institute of Technology (KIT) , 76131 Karlsruhe, Germany
- Physical Institute and Center for Nanotechnology (CeNTech), University of Münster , 48149 Münster, Germany
| | - Michael Hirtz
- Institute of Nanotechnology (INT) and Karlsruhe Nano Micro Facility (KNMF), Karlsruhe Institute of Technology (KIT) , 76131 Karlsruhe, Germany
| |
Collapse
|
44
|
Morena F, Argentati C, Trotta R, Crispoltoni L, Stabile A, Pistilli A, di Baldassarre A, Calafiore R, Montanucci P, Basta G, Pedrinolla A, Smania N, Venturelli M, Schena F, Naro F, Emiliani C, Rende M, Martino S. A Comparison of Lysosomal Enzymes Expression Levels in Peripheral Blood of Mild- and Severe-Alzheimer's Disease and MCI Patients: Implications for Regenerative Medicine Approaches. Int J Mol Sci 2017; 18:ijms18081806. [PMID: 28825628 PMCID: PMC5578193 DOI: 10.3390/ijms18081806] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 08/04/2017] [Accepted: 08/14/2017] [Indexed: 12/22/2022] Open
Abstract
The association of lysosomal dysfunction and neurodegeneration has been documented in several neurodegenerative diseases, including Alzheimer's Disease (AD). Herein, we investigate the association of lysosomal enzymes with AD at different stages of progression of the disease (mild and severe) or with mild cognitive impairment (MCI). We conducted a screening of two classes of lysosomal enzymes: glycohydrolases (β-Hexosaminidase, β-Galctosidase, β-Galactosylcerebrosidase, β-Glucuronidase) and proteases (Cathepsins S, D, B, L) in peripheral blood samples (blood plasma and PBMCs) from mild AD, severe AD, MCI and healthy control subjects. We confirmed the lysosomal dysfunction in severe AD patients and added new findings enhancing the association of abnormal levels of specific lysosomal enzymes with the mild AD or severe AD, and highlighting the difference of AD from MCI. Herein, we showed for the first time the specific alteration of β-Galctosidase (Gal), β-Galactosylcerebrosidase (GALC) in MCI patients. It is notable that in above peripheral biological samples the lysosomes are more sensitive to AD cellular metabolic alteration when compared to levels of Aβ-peptide or Tau proteins, similar in both AD groups analyzed. Collectively, our findings support the role of lysosomal enzymes as potential peripheral molecules that vary with the progression of AD, and make them useful for monitoring regenerative medicine approaches for AD.
Collapse
Affiliation(s)
- Francesco Morena
- Department of Chemistry, Biology and Biotechnology, Biochemistry and Molecular Biology Unit, University of Perugia, Perugia 06123, Italy.
| | - Chiara Argentati
- Department of Chemistry, Biology and Biotechnology, Biochemistry and Molecular Biology Unit, University of Perugia, Perugia 06123, Italy.
| | - Rosa Trotta
- Department of Chemistry, Biology and Biotechnology, Biochemistry and Molecular Biology Unit, University of Perugia, Perugia 06123, Italy.
| | - Lucia Crispoltoni
- Department of Surgery and Biomedical Sciences, Section of Human, Clinical and Forensic Anatomy, School of Medicine, University of Perugia, Perugia 06132, Italy.
| | - Anna Stabile
- Department of Surgery and Biomedical Sciences, Section of Human, Clinical and Forensic Anatomy, School of Medicine, University of Perugia, Perugia 06132, Italy.
| | - Alessandra Pistilli
- Department of Surgery and Biomedical Sciences, Section of Human, Clinical and Forensic Anatomy, School of Medicine, University of Perugia, Perugia 06132, Italy.
| | - Angela di Baldassarre
- Department of Aging Medical Science, University of G. d'Annunzio, Chieti e Pescara, Chieti 66100, Italy.
| | - Riccardo Calafiore
- Department of Medicine, Section of Cardiovascular, Endocrine and Metabolic Clinical Physiology and Laboratory for Endocrine Cell Transplants and Bio-hybrid Organs, University of Perugia, Perugia 06132, Italy.
| | - Pia Montanucci
- Department of Medicine, Section of Cardiovascular, Endocrine and Metabolic Clinical Physiology and Laboratory for Endocrine Cell Transplants and Bio-hybrid Organs, University of Perugia, Perugia 06132, Italy.
| | - Giuseppe Basta
- Department of Medicine, Section of Cardiovascular, Endocrine and Metabolic Clinical Physiology and Laboratory for Endocrine Cell Transplants and Bio-hybrid Organs, University of Perugia, Perugia 06132, Italy.
| | - Anna Pedrinolla
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona 37134, Italy.
| | - Nicola Smania
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona 37134, Italy.
| | - Massimo Venturelli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona 37134, Italy.
| | - Federico Schena
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona 37134, Italy.
| | - Fabio Naro
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Roma, Roma 06100, Italy.
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnology, Biochemistry and Molecular Biology Unit, University of Perugia, Perugia 06123, Italy.
| | - Mario Rende
- Department of Surgery and Biomedical Sciences, Section of Human, Clinical and Forensic Anatomy, School of Medicine, University of Perugia, Perugia 06132, Italy.
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnology, Biochemistry and Molecular Biology Unit, University of Perugia, Perugia 06123, Italy.
| |
Collapse
|
45
|
Rissanou AN, Papananou H, Petrakis VS, Doxastakis M, Andrikopoulos KS, Voyiatzis GA, Chrissopoulou K, Harmandaris V, Anastasiadis SH. Structural and Conformational Properties of Poly(ethylene oxide)/Silica Nanocomposites: Effect of Confinement. Macromolecules 2017. [DOI: 10.1021/acs.macromol.7b00811] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
| | - Hellen Papananou
- Institute
of Electronic Structure and Laser, Foundation for Research and Technology - Hellas,
P.O. Box 1527, 711 10 Heraklion, Crete, Greece
| | | | - Manolis Doxastakis
- Department
of Chemical Engineering, University of Tennessee, Knoxville, Tennessee 37996, USA
| | - Konstantinos S. Andrikopoulos
- Institute
of Chemical Engineering Sciences, Foundation for Research and Technology - Hellas,
P.O. Box 1414, 265 04 Patras, Greece
| | - George A. Voyiatzis
- Institute
of Chemical Engineering Sciences, Foundation for Research and Technology - Hellas,
P.O. Box 1414, 265 04 Patras, Greece
| | - Kiriaki Chrissopoulou
- Institute
of Electronic Structure and Laser, Foundation for Research and Technology - Hellas,
P.O. Box 1527, 711 10 Heraklion, Crete, Greece
| | - Vagelis Harmandaris
- Institute
of Applied and Computational Mathematics, Foundation for Research and Technology - Hellas, P.O. Box 1385, 711 10 Heraklion, Crete, Greece
| | - Spiros H. Anastasiadis
- Institute
of Electronic Structure and Laser, Foundation for Research and Technology - Hellas,
P.O. Box 1527, 711 10 Heraklion, Crete, Greece
| |
Collapse
|
46
|
Wei Y, Mo X, Zhang P, Li Y, Liao J, Li Y, Zhang J, Ning C, Wang S, Deng X, Jiang L. Directing Stem Cell Differentiation via Electrochemical Reversible Switching between Nanotubes and Nanotips of Polypyrrole Array. ACS NANO 2017; 11:5915-5924. [PMID: 28587445 DOI: 10.1021/acsnano.7b01661] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Control of stem cell behaviors at solid biointerfaces is critical for stem-cell-based regeneration and generally achieved by engineering chemical composition, topography, and stiffness. However, the influence of dynamic stimuli at the nanoscale from solid biointerfaces on stem cell fate remains unclear. Herein, we show that electrochemical switching of a polypyrrole (Ppy) array between nanotubes and nanotips can alter surface adhesion, which can strongly influence mechanotransduction activation and guide differentiation of mesenchymal stem cells (MSCs). The Ppy array, prepared via template-free electrochemical polymerization, can be reversibly switched between highly adhesive hydrophobic nanotubes and poorly adhesive hydrophilic nanotips through an electrochemical oxidation/reduction process, resulting in dynamic attachment and detachment to MSCs at the nanoscale. Multicyclic attachment/detachment of the Ppy array to MSCs can activate intracellular mechanotransduction and osteogenic differentiation independent of surface stiffness and chemical induction. This smart surface, permitting transduction of nanoscaled dynamic physical inputs into biological outputs, provides an alternative to classical cell culture substrates for regulating stem cell fate commitment. This study represents a general strategy to explore nanoscaled interactions between stem cells and stimuli-responsive surfaces.
Collapse
Affiliation(s)
- Yan Wei
- Department of Geriatric Dentistry, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Peking University , Beijing 100081, China
| | - Xiaoju Mo
- Department of Geriatric Dentistry, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Peking University , Beijing 100081, China
| | - Pengchao Zhang
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, CAS Center for Excellence in Nanoscience, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences , Beijing 100190, China
- School of Future Technology, University of Chinese Academy of Sciences , Beijing 101408, China
| | - Yingying Li
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, CAS Center for Excellence in Nanoscience, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences , Beijing 100190, China
- School of Future Technology, University of Chinese Academy of Sciences , Beijing 101408, China
| | - Jingwen Liao
- School of Materials Science and Engineering, South China University of Technology , Guangzhou 510641, China
| | - Yongjun Li
- Department of Physics, Beijing Normal University , Beijing 100875, China
| | - Jinxing Zhang
- Department of Physics, Beijing Normal University , Beijing 100875, China
| | - Chengyun Ning
- School of Materials Science and Engineering, South China University of Technology , Guangzhou 510641, China
| | - Shutao Wang
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, CAS Center for Excellence in Nanoscience, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences , Beijing 100190, China
- School of Future Technology, University of Chinese Academy of Sciences , Beijing 101408, China
| | - Xuliang Deng
- Department of Geriatric Dentistry, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Peking University , Beijing 100081, China
| | - Lei Jiang
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, CAS Center for Excellence in Nanoscience, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences , Beijing 100190, China
- School of Future Technology, University of Chinese Academy of Sciences , Beijing 101408, China
| |
Collapse
|
47
|
Vikram Singh A, Gharat T, Batuwangala M, Park B, Endlein T, Sitti M. Three‐dimensional patterning in biomedicine: Importance and applications in neuropharmacology. J Biomed Mater Res B Appl Biomater 2017; 106:1369-1382. [DOI: 10.1002/jbm.b.33922] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 04/19/2017] [Accepted: 04/22/2017] [Indexed: 12/18/2022]
Affiliation(s)
- Ajay Vikram Singh
- Department of Physical IntelligenceMax Planck Institute for Intelligent Systems, Heisenbergstr 370569Stuttgart Germany
| | - Tanmay Gharat
- Department of Chemical and Biological EngineeringRensselaer Polytechnic InstituteNew York New York12180
| | - Madu Batuwangala
- Department of Physical IntelligenceMax Planck Institute for Intelligent Systems, Heisenbergstr 370569Stuttgart Germany
| | - Byung‐Wook Park
- Department of Physical IntelligenceMax Planck Institute for Intelligent Systems, Heisenbergstr 370569Stuttgart Germany
| | - Thomas Endlein
- Department of Physical IntelligenceMax Planck Institute for Intelligent Systems, Heisenbergstr 370569Stuttgart Germany
| | - Metin Sitti
- Department of Physical IntelligenceMax Planck Institute for Intelligent Systems, Heisenbergstr 370569Stuttgart Germany
| |
Collapse
|
48
|
Wei M, Li S, Yang Z, Zheng W, Le W. Gold nanoparticles enhance the differentiation of embryonic stem cells into dopaminergic neurons via mTOR/p70S6K pathway. Nanomedicine (Lond) 2017; 12:1305-1317. [PMID: 28520507 DOI: 10.2217/nnm-2017-0001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 03/17/2017] [Indexed: 01/12/2023] Open
Abstract
AIM This study aimed to investigate the effect of gold nanoparticles (AuNPs) on differentiation of mouse embryonic stem cells (ESCs) into dopaminergic (DA) neurons and explore the possible underlying molecular mechanisms. MATERIALS & METHODS The efficiency of AuNPs on DA neuron differentiation was evaluated by observing fluorescence in TH promoter-engineered GFP-reporter ESCs, western blot and real-time PCR. The possible signal pathway was determined by western blot. RESULTS Compared with feeder-free control condition, AuNPs are able to enhance fate specification of ESCs into DA neurons. Moreover, mTOR/p70S6K signaling pathway was found involving in this AuNPs-mediated DA neuron differentiation. CONCLUSION Our findings may lead future insight investigation into the underlying mechanisms and potential application of AuNPs in stem cell research.
Collapse
Affiliation(s)
- Min Wei
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Song Li
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Zhaofei Yang
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Wei Zheng
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Weidong Le
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
49
|
Neuroregeneration versus neurodegeneration: toward a paradigm shift in Alzheimer's disease drug discovery. Future Med Chem 2017. [DOI: 10.4155/fmc-2017-0038] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Alzheimer's disease represents an enormous global burden in terms of human suffering and economic cost. To tackle the current lack of effective drugs and the continuous clinical trial failures might require a shift from the prevailing paradigm targeting pathogenesis to the one targeting neural stem cells (NSCs) regeneration. In this context, small molecules have come to the forefront for their potential to manipulate NSCs, provide therapeutic tools and unveil NSCs biology. Classically, these molecules have been generated either by target-based or phenotypic approaches. To circumvent specific liabilities, nanomedicines emerge as a feasible alternative. However, this review is not intended to be comprehensive. Its purpose is to focus on recent examples that could accelerate development of neuroregenerative drugs against Alzheimer's disease.
Collapse
|
50
|
Merolli A, Mao Y, Kohn J. A suspended carbon fiber culture to model myelination by human Schwann cells. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2017; 28:57. [PMID: 28210970 DOI: 10.1007/s10856-017-5867-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 02/07/2017] [Indexed: 06/06/2023]
Abstract
Understanding of myelination/remyelination process is essential to guide tissue engineering for nerve regeneration. In vitro models currently used are limited to cell population studies and cannot easily identify individual cell contribution to the process. We established a novel model to study the contribution of human Schwann cells to the myelination process. The model avoids the presence of neurons in culture; Schwann cells respond solely to the biophysical properties of an artificial axon. The model uses a single carbon fiber suspended in culture media far from the floor of the well. The fiber provides an elongated structure of defined diameter with 360-degree of surface available for human Schwann cells to wrap around. This model enabled us to spatially and temporally track the myelination by individual Schwann cells along the fiber. We observed cell attachment, elongation and wrapping over a period of 9 days. Cells remained alive and expressed Myelin Basic Protein and Myelin Associated Glycoprotein as expected. Natural and artificial molecules, and external physical factors (e.g., p atterned electrical impulses), may be tested with this model as possible regulators of myelination.
Collapse
Affiliation(s)
- Antonio Merolli
- New Jersey Center for Biomaterials, Rutgers-The State University of New Jersey, 145 Bevier Rd., Piscataway, NJ, 08854, USA.
- Policlinico Gemelli, Universita' Cattolica del Sacro Cuore, largo Gemelli 8, 00168, Rome, Italy.
| | - Yong Mao
- New Jersey Center for Biomaterials, Rutgers-The State University of New Jersey, 145 Bevier Rd., Piscataway, NJ, 08854, USA
| | - Joachim Kohn
- New Jersey Center for Biomaterials, Rutgers-The State University of New Jersey, 145 Bevier Rd., Piscataway, NJ, 08854, USA
| |
Collapse
|