1001
|
Zhang J, Mou L, Jiang X. Hydrogels Incorporating Au@Polydopamine Nanoparticles: Robust Performance for Optical Sensing. Anal Chem 2018; 90:11423-11430. [PMID: 30191718 DOI: 10.1021/acs.analchem.8b02459] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Stimuli-responsive hydrogels (SRhG) that undergo response to physicochemical stimuli have been broadly applied in separation, biosensing, and drug delivery. Since, most of the SRhG are based on the structural behaviors (swelling or collapse). Herein, we describe a more simple and convenient colorimetric SRhG of polydopamine-coated gold nanoparticles (Au@PDA NPs) hydrogel. The newly developed SRhG is based on the in situ surface chemistry of Au@PDA NPs with core-shell structure embedding in agarose hydrogel. Silver ions can in situ form Ag NPs on surfaces of Au@PDA NPs (Ag_Au@PDA NPs with core-satellites like structure) at ambient conditions, which shift the localized surface plasmon resonance (LSPR) absorption peak and result in color change. The solid sensing phase of SRhG shows greatly improved stability and anti-interference ability comparing to that of solution phase sensing. With rational designs, Au@PDA NPs hydrogel shows great potential in optical sensing, for example, biothiol detection, and coupled with enzyme-cascade reaction for acetylcholinesterase activity detection and inhibitor assays with excellent sensitivity and selectivity.
Collapse
Affiliation(s)
- Jiangjiang Zhang
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology , Number 11 Zhongguancun Beiyitiao , Beijing 100190 , China.,Sino-Danish College , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Lei Mou
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology , Number 11 Zhongguancun Beiyitiao , Beijing 100190 , China.,Academy for Advanced Interdisciplinary Studies , Peking University , Beijing 100871 , China
| | - Xingyu Jiang
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology , Number 11 Zhongguancun Beiyitiao , Beijing 100190 , China.,Sino-Danish College , University of Chinese Academy of Sciences , Beijing 100049 , China
| |
Collapse
|
1002
|
Youngblood RL, Truong NF, Segura T, Shea LD. It's All in the Delivery: Designing Hydrogels for Cell and Non-viral Gene Therapies. Mol Ther 2018; 26:2087-2106. [PMID: 30107997 PMCID: PMC6127639 DOI: 10.1016/j.ymthe.2018.07.022] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 07/24/2018] [Accepted: 07/24/2018] [Indexed: 01/08/2023] Open
Abstract
Hydrogels provide a regenerative medicine platform with their ability to create an environment that supports transplanted or endogenous infiltrating cells and enables these cells to restore or replace the function of tissues lost to disease or trauma. Furthermore, these systems have been employed as delivery vehicles for therapeutic genes, which can direct and/or enhance the function of the transplanted or endogenous cells. Herein, we review recent advances in the development of hydrogels for cell and non-viral gene delivery through understanding the design parameters, including both physical and biological components, on promoting transgene expression, cell engraftment, and ultimately cell function. Furthermore, this review identifies emerging opportunities for combining cell and gene delivery approaches to overcome challenges to the field.
Collapse
Affiliation(s)
- Richard L Youngblood
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Norman F Truong
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tatiana Segura
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA.
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
1003
|
Rajkovic O, Potjewyd G, Pinteaux E. Regenerative Medicine Therapies for Targeting Neuroinflammation After Stroke. Front Neurol 2018; 9:734. [PMID: 30233484 PMCID: PMC6129611 DOI: 10.3389/fneur.2018.00734] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/13/2018] [Indexed: 12/15/2022] Open
Abstract
Inflammation is a major pathological event following ischemic stroke that contributes to secondary brain tissue damage leading to poor functional recovery. Following the initial ischemic insult, post-stroke inflammatory damage is driven by initiation of a central and peripheral innate immune response and disruption of the blood-brain barrier (BBB), both of which are triggered by the release of pro-inflammatory cytokines and infiltration of circulating immune cells. Stroke therapies are limited to early cerebral blood flow reperfusion, and whilst current strategies aim at targeting neurodegeneration and/or neuroinflammation, innovative research in the field of regenerative medicine aims at developing effective treatments that target both the acute and chronic phase of inflammation. Anti-inflammatory regenerative strategies include the use of nanoparticles and hydrogels, proposed as therapeutic agents and as a delivery vehicle for encapsulated therapeutic biological factors, anti-inflammatory drugs, stem cells, and gene therapies. Biomaterial strategies-through nanoparticles and hydrogels-enable the administration of treatments that can more effectively cross the BBB when injected systemically, can be injected directly into the brain, and can be 3D-bioprinted to create bespoke implants within the site of ischemic injury. In this review, these emerging regenerative and anti-inflammatory approaches will be discussed in relation to ischemic stroke, with a perspective on the future of stroke therapies.
Collapse
Affiliation(s)
- Olivera Rajkovic
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Geoffrey Potjewyd
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Emmanuel Pinteaux
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
1004
|
Macdougall LJ, Wiley KL, Kloxin AM, Dove AP. Design of synthetic extracellular matrices for probing breast cancer cell growth using robust cyctocompatible nucleophilic thiol-yne addition chemistry. Biomaterials 2018; 178:435-447. [PMID: 29773227 PMCID: PMC6699181 DOI: 10.1016/j.biomaterials.2018.04.046] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/27/2018] [Accepted: 04/23/2018] [Indexed: 12/16/2022]
Abstract
Controlled, three-dimensional (3D) cell culture systems are of growing interest for both tissue regeneration and disease, including cancer, enabling hypothesis testing about the effects of microenvironment cues on a variety of cellular processes, including aspects of disease progression. In this work, we encapsulate and culture in three dimensions different cancer cell lines in a synthetic extracellular matrix (ECM), using mild and efficient chemistry. Specifically, harnessing the nucleophilic addition of thiols to activated alkynes, we have created hydrogel-based materials with multifunctional poly(ethylene glycol) (PEG) and select biomimetic peptides. These materials have definable, controlled mechanical properties (G' = 4-10 kPa) and enable facile incorporation of pendant peptides for cell adhesion, relevant for mimicking soft tissues, where polymer architecture allows tuning of matrix degradation. These matrices rapidly formed in the presence of sensitive breast cancer cells (MCF-7) for successful encapsulation with high cell viability, greatly improved relative to that observed with the more widely used radically-initiated thiol-ene crosslinking chemistry. Furthermore, controlled matrix degradation by both bulk and local mechanisms, ester hydrolysis of the polymer network and cell-driven enzymatic hydrolysis of cell-degradable peptide, allowed cell proliferation and the formation of cell clusters within these thiol-yne hydrogels. These studies demonstrate the importance of chemistry in ECM mimics and the potential thiol-yne chemistry has as a crosslinking reaction for the encapsulation and culture of cells, including those sensitive to radical crosslinking pathways.
Collapse
Affiliation(s)
- Laura J Macdougall
- Department of Chemistry, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Katherine L Wiley
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
| | - April M Kloxin
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA; Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
| | - Andrew P Dove
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
1005
|
Wu S, Yue H, Wu J, Zhang W, Jiang M, Ma G. The interacting role of physical stiffness and tumor cells on the macrophages polarization. Colloids Surf A Physicochem Eng Asp 2018. [DOI: 10.1016/j.colsurfa.2018.04.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
1006
|
Heidari H, Taylor H. Review Article: Capturing the physiological complexity of the brain's neuro-vascular unit in vitro. BIOMICROFLUIDICS 2018; 12:051502. [PMID: 30364144 PMCID: PMC6191301 DOI: 10.1063/1.5045126] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 09/27/2018] [Indexed: 06/01/2023]
Abstract
With the accelerating pace of brain research in recent years and the growing appreciation of the complexity of the brain and several brain-associated neurological diseases, the demand for powerful tools to enhance drug screening, diagnosis, and fundamental research is greater than ever. Highly representative models of the central nervous system (CNS) can play a critical role in meeting these needs. Unfortunately, in vivo animal models lack controllability, are difficult to monitor, and do not model human-specific brain behavior accurately. On the other hand, in silico computational models struggle to capture comprehensively the intertwined biological, chemical, electrical, and mechanical complexity of the brain. This leaves us with the promising domain of "organ-on-chip" in vitro models. In this review, we describe some of the most pioneering efforts in this expanding field, offering a perspective on the new possibilities as well as the limitations of each approach. We focus particularly on how the models reproduce the blood-brain barrier (BBB), which mediates mass transport to and from brain tissue. We also offer a brief commentary on strategies for evaluating the blood-brain barrier functionality of these in vitro models, including trans-endothelial electrical resistance (TEER), immunocytochemistry, and permeability analysis. From the early membrane-based models of the BBB that have grown into the Transwell® class of devices, to the era of microfluidic chips and a future of bio-printed tissue, we see enormous improvement in the reliability of in vitro models. More and more of the biological and structural complexity of the BBB is being captured by microfluidic chips, and the organ-specificity of bio-printed tissue is also significantly improved. Although we believe that the long-term solution will eventually take the form of automated and parallelized bio-printing systems, we find that valuable transport studies can already be accomplished with microfluidic platforms.
Collapse
Affiliation(s)
- Hossein Heidari
- Department of Mechanical Engineering, University of California, 6159 Etcheverry Hall, Berkeley, California 94720, USA
| | - Hayden Taylor
- Department of Mechanical Engineering, University of California, 6159 Etcheverry Hall, Berkeley, California 94720, USA
| |
Collapse
|
1007
|
Bahmad HF, Cheaito K, Chalhoub RM, Hadadeh O, Monzer A, Ballout F, El-Hajj A, Mukherji D, Liu YN, Daoud G, Abou-Kheir W. Sphere-Formation Assay: Three-Dimensional in vitro Culturing of Prostate Cancer Stem/Progenitor Sphere-Forming Cells. Front Oncol 2018; 8:347. [PMID: 30211124 PMCID: PMC6121836 DOI: 10.3389/fonc.2018.00347] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/07/2018] [Indexed: 12/12/2022] Open
Abstract
Cancer Stem Cells (CSCs) are a sub-population of cells, identified in most tumors, responsible for the initiation, recurrence, metastatic potential, and resistance of different malignancies. In prostate cancer (PCa), CSCs were identified and thought to be responsible for the generation of the lethal subtype, commonly known as Castration-Resistant Prostate Cancer (CRPC). In vitro models to investigate the properties of CSCs in PCa are highly required. Sphere-formation assay is an in vitro method commonly used to identify CSCs and study their properties. Here, we report the detailed methodology on how to generate and propagate spheres from PCa cell lines and from murine prostate tissue. This model is based on the ability of stem cells to grow in non-adherent serum-free gel matrix. We also describe how to use these spheres in histological and immuno-fluorescent staining assays to assess the differentiation potential of the CSCs. Our results show the sphere-formation Assay (SFA) as a reliable in vitro assay to assess the presence and self-renewal ability of CSCs in different PCa models. This platform presents a useful tool to evaluate the effect of conventional or novel agents on the initiation and self-renewing properties of different tumors. The effects can be directly evaluated through assessment of the sphere-forming efficiency (SFE) over five generations or other downstream assays such as immuno-histochemical analysis of the generated spheres.
Collapse
Affiliation(s)
- Hisham F Bahmad
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Katia Cheaito
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Reda M Chalhoub
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ola Hadadeh
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Alissar Monzer
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Farah Ballout
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Beirut, Lebanon
| | - Albert El-Hajj
- Division of Urology, Department of Surgery, American University of Beirut Medical Center, Beirut, Lebanon
| | - Deborah Mukherji
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Yen-Nien Liu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Georges Daoud
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
1008
|
Wang S, Mao S, Li M, Li HF, Lin JM. Near-physiological microenvironment simulation on chip to evaluate drug resistance of different loci in tumour mass. Talanta 2018; 191:67-73. [PMID: 30262100 DOI: 10.1016/j.talanta.2018.08.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/27/2018] [Accepted: 08/03/2018] [Indexed: 12/17/2022]
Abstract
Developing a bio-functional model in vitro to study cancer resistance, which is a big challenge for clinical cancer therapy, is of great interest. Such reliable model requires appropriate drug diffusion kinetics simulation and a microenvironment that allows cell-cell and cell-matrix interactions. In this work, a special hydrogel-based three-dimensional (3D) microfluidic chip was constructed to simulate tumour-vascular microenvironment. The self-healing hydrogel supports long-time cell survival and proliferation, effective cellular metabolism of cancer drugs and cell-cell interaction between different types of cells. In the effective near-physiological tumour-vascular microenvironment, the endothelial and fibroblast cells are spread on different sides of a porous membrane, while sensitive and resistant breast tumour cells are separately cultured in the dynamic hydrogel consisting of glycol chitosan and telechelic difunctional poly (ethylene glycol) in the upper chambers. Nutrients and drugs are introduced through the bottom channel and diffuse into the cancer cells. Doxorubicin molecules pass first through blood vessel endothelial cells and act on the tumour cells surrounded by fibroblasts. Tumour cells respond differently to drug when they are cultured in the microenvironment. Sensitive breast tumour cells have a 47% increase in viability than those cultured without fibroblasts and endothelial cells. Both sensitive and resistant tumour cells can be analysed under the same chemical environment. This work represents a multi-functional in vitro platform that allows near-physiological simulation, effective drug metabolism and cellular response to extracellular stimuli and has great potential to make drug discovery speedy and precise.
Collapse
Affiliation(s)
- Shiqi Wang
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Sifeng Mao
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Min Li
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Hai-Fang Li
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China.
| | - Jin-Ming Lin
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
1009
|
Paul W, Marta C, Tom VDW. Resolving host–microbe interactions in the gut: the promise of in vitro models to complement in vivo research. Curr Opin Microbiol 2018; 44:28-33. [DOI: 10.1016/j.mib.2018.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 06/26/2018] [Accepted: 07/03/2018] [Indexed: 12/17/2022]
|
1010
|
Shi P, Kim YH, Mousa M, Sanchez RR, Oreffo ROC, Dawson JI. Self-Assembling Nanoclay Diffusion Gels for Bioactive Osteogenic Microenvironments. Adv Healthc Mater 2018; 7:e1800331. [PMID: 29911340 DOI: 10.1002/adhm.201800331] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Indexed: 11/08/2022]
Abstract
Laponite nanoparticles have attracted attention in the tissue engineering field for their protein interactions, gel-forming properties, and, more recently, osteogenic bioactivity. Despite growing interest in the osteogenic properties of Laponite, the application of Laponite colloidal gels to host the osteogenic differentiation of responsive stem cell populations remains unexplored. Here, the potential to harness the gel-forming properties of Laponite to generate injectable bioactive microenvironments for osteogenesis is demonstrated. A diffusion/dialysis gelation method allows the rapid formation of stable transparent gels from injectable, thixotropic Laponite suspensions in physiological fluids. Upon contact with buffered saline or blood serum, nanoporous gel networks exhibiting, respectively, fivefold and tenfold increases in gel stiffness are formed due to the reorganization of nanoparticle interactions. Laponite diffusion gels are explored as osteogenic microenvironments for skeletal stem cell containing populations. Laponite films support cell adhesion, proliferation, and differentiation of human bone marrow stromal cells in 2D. Laponite gel encapsulation significantly enhances osteogenic protein expression compared with 3D pellet culture controls. In both 2D and 3D conditions, cell associated mineralization is strongly enhanced. This study demonstrates that Laponite diffusion gels offer considerable potential as biologically active and clinically relevant bone tissue engineering scaffolds.
Collapse
Affiliation(s)
- Pujiang Shi
- Bone and Joint Research Group; Centre for Human Development; Stem Cells and Regeneration; Institute of Developmental Sciences; University of Southampton; Southampton SO16 6YD UK
| | - Yang-Hee Kim
- Bone and Joint Research Group; Centre for Human Development; Stem Cells and Regeneration; Institute of Developmental Sciences; University of Southampton; Southampton SO16 6YD UK
| | - Mohamed Mousa
- Bone and Joint Research Group; Centre for Human Development; Stem Cells and Regeneration; Institute of Developmental Sciences; University of Southampton; Southampton SO16 6YD UK
| | - Roxanna Ramnarine Sanchez
- Bone and Joint Research Group; Centre for Human Development; Stem Cells and Regeneration; Institute of Developmental Sciences; University of Southampton; Southampton SO16 6YD UK
| | - Richard O. C. Oreffo
- Bone and Joint Research Group; Centre for Human Development; Stem Cells and Regeneration; Institute of Developmental Sciences; University of Southampton; Southampton SO16 6YD UK
| | - Jonathan I. Dawson
- Bone and Joint Research Group; Centre for Human Development; Stem Cells and Regeneration; Institute of Developmental Sciences; University of Southampton; Southampton SO16 6YD UK
| |
Collapse
|
1011
|
Bao M, Xie J, Huck WTS. Recent Advances in Engineering the Stem Cell Microniche in 3D. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1800448. [PMID: 30128252 PMCID: PMC6096985 DOI: 10.1002/advs.201800448] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/01/2018] [Indexed: 05/18/2023]
Abstract
Conventional 2D cell culture techniques have provided fundamental insights into key biochemical and biophysical mechanisms responsible for various cellular behaviors, such as cell adhesion, spreading, division, proliferation, and differentiation. However, 2D culture in vitro does not fully capture the physical and chemical properties of the native microenvironment. There is a growing body of research that suggests that cells cultured on 2D substrates differ greatly from those grown in vivo. This article focuses on recent progress in using bioinspired 3D matrices that recapitulate as many aspects of the natural extracellular matrix as possible. A range of techniques for the engineering of 3D microenvironment with precisely controlled biophysical and chemical properties, and the impact of these environments on cellular behavior, is reviewed. Finally, an outlook on future challenges for engineering the 3D microenvironment and how such approaches would further our understanding of the influence of the microenvironment on cell function is provided.
Collapse
Affiliation(s)
- Min Bao
- Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 1356525 AJNijmegenThe Netherlands
| | - Jing Xie
- Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 1356525 AJNijmegenThe Netherlands
| | - Wilhelm T. S. Huck
- Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 1356525 AJNijmegenThe Netherlands
| |
Collapse
|
1012
|
Zhou X, Li Z. Advances and Biomedical Applications of Polypeptide Hydrogels Derived from α-Amino Acid N-Carboxyanhydride (NCA) Polymerizations. Adv Healthc Mater 2018; 7:e1800020. [PMID: 29869375 DOI: 10.1002/adhm.201800020] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/18/2018] [Indexed: 02/06/2023]
Abstract
Polypeptide hydrogels, having the ability to mimic certain properties of natural, native extracellular matrix components, are being actively designed and described for various applications in the construction of tissue engineering scaffolds, living cell encapsulation, and drug delivery systems. Compared to conventional hydrogels, polypeptide hydrogels possess biocompatibility, biodegradability, bioactivity, functional diversity, and structural advantage based on the unique secondary structures (α-helix and β-sheet). Furthermore, the progresses in functional N-carboxyanhydride polymerization combined with advanced orthogonal conjugation techniques significantly promote the development of the polypeptide materials. This progress report focuses on the recent advances in designing and engineering polypeptide hydrogels obtained from ring opening polymerization, highlighting the precise manipulation of their properties for biomedical applications.
Collapse
Affiliation(s)
- Xianfeng Zhou
- Key Lab of Biobased Polymer Materials of Shandong Provincial Education Department; School of Polymer Science and Engineering; Qingdao University of Science and Technology; Qingdao 266042 China
- Department of Polymer Science; University of Akron; Akron OH 44325 USA
| | - Zhibo Li
- Key Lab of Biobased Polymer Materials of Shandong Provincial Education Department; School of Polymer Science and Engineering; Qingdao University of Science and Technology; Qingdao 266042 China
| |
Collapse
|
1013
|
Tharp KM, Weaver VM. Modeling Tissue Polarity in Context. J Mol Biol 2018; 430:3613-3628. [PMID: 30055167 DOI: 10.1016/j.jmb.2018.07.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 06/27/2018] [Accepted: 07/11/2018] [Indexed: 12/17/2022]
Abstract
Polarity is critical for development and tissue-specific function. However, the acquisition and maintenance of tissue polarity is context dependent. Thus, cell and tissue polarity depend on cell adhesion which is regulated by the cytoskeleton and influenced by the biochemical composition of the extracellular microenvironment and modified by biomechanical cues within the tissue. These biomechanical cues include fluid flow induced shear stresses, cell-density and confinement-mediated compression, and cellular actomyosin tension intrinsic to the tissue or induced in response to morphogens or extracellular matrix stiffness. Here, we discuss how extracellular matrix stiffness and fluid flow influence cell-cell and cell-extracellular matrix adhesion and alter cytoskeletal organization to modulate cell and tissue polarity. We describe model systems that when combined with state of the art molecular screens and high-resolution imaging can be used to investigate how force modulates cell and tissue polarity.
Collapse
Affiliation(s)
- Kevin M Tharp
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA; Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94143, USA; Department of Radiation Oncology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
1014
|
Tuneable hydrogels of Caf1 protein fibers. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 93:88-95. [PMID: 30274124 DOI: 10.1016/j.msec.2018.07.063] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 07/12/2018] [Accepted: 07/23/2018] [Indexed: 01/20/2023]
Abstract
Capsular antigen fraction 1 (Caf1) is a robust polymeric protein forming a protective layer around the bacterium Yersinia pestis. Occurring as ≈1 μm polymeric fibers, it shares its immunoglobulin-like fold with the majority of mammalian extracellular proteins such as fibronectin and this structural similarity suggests that this unusual polymer could form useful mimics of the extracellular matrix. Driven by the pressing need for reliable animal-free 3D cell culture environments, we showed previously that recombinant Caf1 produced in Escherichia coli can be engineered to include bioactive peptides, which influence cell behavior. Here, we demonstrate that through chemical crosslinking with a small palette of PEG-based crosslinkers, Caf1-based hydrogels can be prepared displaying a wide range of mechanical and morphological properties that were studied by rheology, compressive testing, SDS-PAGE and scanning electron microscopy. By varying the Caf1 protein concentration, viscoelasticity and stiffness (~11-2300 Pa) are reproducibly tunable to match natural and commercial 3D gels. Hydrogel porosity and swelling ratios were found to be defined by crosslinker architecture and concentration. Finally the hydrogels, which are 95-99% water, were shown to retain the high stability of the native Caf1 protein in a range of aqueous conditions, including extended immersion in cell culture media. The unusual Caf1 polymer thus offers the possibility of presenting bioactive protein subunits in a precisely tuneable hydrogel for use in cell culture and drug delivery applications.
Collapse
|
1015
|
Burek M, Waśkiewicz S, Lalik A, Student S, Bieg T, Wandzik I. Thermoresponsive microgels containing trehalose as soft matrices for 3D cell culture. Biomater Sci 2018; 5:234-246. [PMID: 27921099 DOI: 10.1039/c6bm00624h] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A series of thermoresponsive glycomicrogels with trehalose in the cross-links or with trehalose in the cross-links and as pending moieties was synthesized. These materials were obtained by surfactant-free precipitation copolymerization of N-isopropylacrylamide and various amounts of trehalose monomers. The resultant particles showed a spherical shape and a submicrometer hydrodynamic size with a narrow size distribution. At 25 °C, glycomicrogels in solutions with physiological ionic strength formed stable colloids, which further gelled upon heating to physiological temperature forming a macroscopic hydrogel with an interconnected porous structure. These extremely soft matrices with dynamic storage modulus in the range of 9-70 Pa were examined in 3D culture systems for HeLa cell culture in comparison to traditional 2D mode. They showed relatively low syneresis over time, especially when glycomicrogels with a high content of hydrophilic trehalose were used as building blocks. An incorporated pending trehalose composed of two α,α'-1,1'-linked d-glucose moieties was used with the intention of providing multivalent interactions with glucose transporters (GLUTs) expressed on the cell surface. A better cell viability was observed when a soft hydrogel with the highest content of trehalose and the lowest syneresis was used as a matrix compared to a 2D control assay.
Collapse
Affiliation(s)
- Małgorzata Burek
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Faculty of Chemistry, Silesian University of Technology, B. Krzywoustego 4, 44 100 Gliwice, Poland.
| | - Sylwia Waśkiewicz
- Department of Physical Chemistry and Technology of Polymers, Faculty of Chemistry, Silesian University of Technology, M. Strzody 9, 44 100 Gliwice, Poland
| | - Anna Lalik
- Systems Engineering Group, Institute of Automatic Control, Silesian University of Technology, B. Krzywoustego 8, 44 100 Gliwice, Poland
| | - Sebastian Student
- Systems Engineering Group, Institute of Automatic Control, Silesian University of Technology, B. Krzywoustego 8, 44 100 Gliwice, Poland
| | - Tadeusz Bieg
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Faculty of Chemistry, Silesian University of Technology, B. Krzywoustego 4, 44 100 Gliwice, Poland.
| | - Ilona Wandzik
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Faculty of Chemistry, Silesian University of Technology, B. Krzywoustego 4, 44 100 Gliwice, Poland.
| |
Collapse
|
1016
|
Ferreira LP, Gaspar VM, Mano JF. Design of spherically structured 3D in vitro tumor models -Advances and prospects. Acta Biomater 2018; 75:11-34. [PMID: 29803007 DOI: 10.1016/j.actbio.2018.05.034] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 05/17/2018] [Accepted: 05/22/2018] [Indexed: 12/29/2022]
Abstract
Three-dimensional multicellular tumor models are receiving an ever-growing focus as preclinical drug-screening platforms due to their potential to recapitulate major physiological features of human tumors in vitro. In line with this momentum, the technologies for assembly of 3D microtumors are rapidly evolving towards a comprehensive inclusion of tumor microenvironment elements. Customized spherically structured platforms, including microparticles and microcapsules, provide a robust and scalable technology to imprint unique biomolecular tumor microenvironment hallmarks into 3D in vitro models. Herein, a comprehensive overview of novel advances on the integration of tumor-ECM components and biomechanical cues into 3D in vitro models assembled in spherical shaped platforms is provided. Future improvements regarding spatiotemporal/mechanical adaptability, and degradability, during microtumors in vitro 3D culture are also critically discussed considering the realistic potential of these platforms to mimic the dynamic tumor microenvironment. From a global perspective, the production of 3D multicellular spheroids with tumor ECM components included in spherical models will unlock their potential to be used in high-throughput screening of therapeutic compounds. It is envisioned, in a near future, that a combination of spherically structured 3D microtumor models with other advanced microfluidic technologies will properly recapitulate the flow dynamics of human tumors in vitro. STATEMENT OF SIGNIFICANCE The ability to correctly mimic the complexity of the tumor microenvironment in vitro is a key aspect for the development of evermore realistic in vitro models for drug-screening and fundamental cancer biology studies. In this regard, conventional spheroid-based 3D tumor models, combined with spherically structured biomaterials, opens the opportunity to precisely recapitulate complex cell-extracellular matrix interactions and tumor compartmentalization. This review provides an in-depth focus on current developments regarding spherically structured scaffolds engineered into in vitro 3D tumor models, and discusses future advances toward all-encompassing platforms that may provide an improved in vitro/in vivo correlation in a foreseeable future.
Collapse
Affiliation(s)
- L P Ferreira
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - V M Gaspar
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| | - J F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| |
Collapse
|
1017
|
Ma S, Mukherjee N. Microfluidics Fabrication of Soft Microtissues and Bottom-Up Assembly. ACTA ACUST UNITED AC 2018. [DOI: 10.1002/adbi.201800119] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Shaohua Ma
- Tsinghua-Berkeley Shenzhen Institute; Tsinghua University; Shenzhen China
| | - Nobina Mukherjee
- Department of Chemistry; University of Oxford; OX1 3TA Oxford UK
| |
Collapse
|
1018
|
Oliveira MB, Bastos HXS, Mano JF. Sequentially Moldable and Bondable Four-Dimensional Hydrogels Compatible with Cell Encapsulation. Biomacromolecules 2018; 19:2742-2749. [PMID: 29698598 PMCID: PMC6450509 DOI: 10.1021/acs.biomac.8b00337] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Hydrogels have captivated the attention of several research and industry segments, including bioengineering, tissue engineering, implantable/wearable sensors and actuators, bioactive agent delivery, food processing, and industrial processes optimization. A common limitation of these systems is their fixed shape. The concept of hydrogel moldability is often assigned to the injectability potential of liquid precursors, and this feature is often lost right after hydrogel formation. Hydrogel modulation is a recent trend that advocates the importance of designing materials with shape fitting ability targeting on-demand responses or defect filling purposes. Here, we present a compliant and cell encapsulation-compatible hydrogel prepared from unmodified natural origin polymers with the ability to undergo extreme sequential shape alterations with high recovery of its mechanical properties. Different fragments of these hydrogels could be bonded together in spatiotemporally controlled shape- and formulation-morphing structures. This material is prepared with affordable off-the-shelf polysaccharides of natural origin using a mild and safe processing strategy based solely on polyelectrolyte complexation followed by an innovative partial coacervate compaction and dehydration step. These unique hydrogels hold potential for multifield industrial and healthcare applications. In particular, they may find application as defect filling agents or highly compliant wound healing patches for cargo release and/or cell delivery for tissue regeneration and cell-based therapies.
Collapse
Affiliation(s)
- Mariana B. Oliveira
- Department of Chemistry, CICECO – Aveiro Institute of Materials. University of Aveiro. 3810-193 Aveiro, Portugal
| | - Henrique X. S. Bastos
- Department of Chemistry, CICECO – Aveiro Institute of Materials. University of Aveiro. 3810-193 Aveiro, Portugal
| | - João F. Mano
- Department of Chemistry, CICECO – Aveiro Institute of Materials. University of Aveiro. 3810-193 Aveiro, Portugal
| |
Collapse
|
1019
|
Nagao M, Sengupta J, Diaz-Dussan D, Adam M, Wu M, Acker J, Ben R, Ishihara K, Zeng H, Miura Y, Narain R. Synthesis of Highly Biocompatible and Temperature-Responsive Physical Gels for Cryopreservation and 3D Cell Culture. ACS APPLIED BIO MATERIALS 2018; 1:356-366. [DOI: 10.1021/acsabm.8b00096] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Masanori Nagao
- Department of Chemical Engineering, Kyushu University, Fukuoka 819-0395, Japan
| | | | | | - Madeleine Adam
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | | | - Jason Acker
- Centre for Innovation, Canadian Blood Services, Edmonton, Alberta T6G 2R8, Canada
| | - Robert Ben
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Kazuhiko Ishihara
- Department of Materials Engineering, The University of Tokyo, Tokyo 113-8654, Japan
| | | | - Yoshiko Miura
- Department of Chemical Engineering, Kyushu University, Fukuoka 819-0395, Japan
| | | |
Collapse
|
1020
|
Bastounis EE, Ortega FE, Serrano R, Theriot JA. A Multi-well Format Polyacrylamide-based Assay for Studying the Effect of Extracellular Matrix Stiffness on the Bacterial Infection of Adherent Cells. J Vis Exp 2018. [PMID: 30035758 PMCID: PMC6124605 DOI: 10.3791/57361] [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] [Indexed: 01/05/2023] Open
Abstract
Extracellular matrix stiffness comprises one of the multiple environmental mechanical stimuli that are well known to influence cellular behavior, function, and fate in general. Although increasingly more adherent cell types' responses to matrix stiffness have been characterized, how adherent cells' susceptibility to bacterial infection depends on matrix stiffness is largely unknown, as is the effect of bacterial infection on the biomechanics of host cells. We hypothesize that the susceptibility of host endothelial cells to a bacterial infection depends on the stiffness of the matrix on which these cells reside, and that the infection of the host cells with bacteria will change their biomechanics. To test these two hypotheses, endothelial cells were used as model hosts and Listeria monocytogenes as a model pathogen. By developing a novel multi-well format assay, we show that the effect of matrix stiffness on infection of endothelial cells by L. monocytogenes can be quantitatively assessed through flow cytometry and immunostaining followed by microscopy. In addition, using traction force microscopy, the effect of L. monocytogenes infection on host endothelial cell biomechanics can be studied. The proposed method allows for the analysis of the effect of tissue-relevant mechanics on bacterial infection of adherent cells, which is a critical step towards understanding the biomechanical interactions between cells, their extracellular matrix, and pathogenic bacteria. This method is also applicable to a wide variety of other types of studies on cell biomechanics and response to substrate stiffness where it is important to be able to perform many replicates in parallel in each experiment.
Collapse
Affiliation(s)
| | - Fabian E Ortega
- Department of Biochemistry, Stanford University School of Medicine
| | - Ricardo Serrano
- Department of Mechanical and Aerospace Engineering, University of California San Diego
| | - Julie A Theriot
- Departments of Biochemistry, Microbiology and Immunology and Howard Hughes Medical Institute, Stanford University School of Medicine
| |
Collapse
|
1021
|
Jiang T, Munguia-Lopez J, Flores-Torres S, Grant J, Vijayakumar S, De Leon-Rodriguez A, Kinsella JM. Bioprintable Alginate/Gelatin Hydrogel 3D In Vitro Model Systems Induce Cell Spheroid Formation. J Vis Exp 2018. [PMID: 30010644 DOI: 10.3791/57826] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The cellular, biochemical, and biophysical heterogeneity of the native tumor microenvironment is not recapitulated by growing immortalized cancer cell lines using conventional two-dimensional (2D) cell culture. These challenges can be overcome by using bioprinting techniques to build heterogeneous three-dimensional (3D) tumor models whereby different types of cells are embedded. Alginate and gelatin are two of the most common biomaterials employed in bioprinting due to their biocompatibility, biomimicry, and mechanical properties. By combining the two polymers, we achieved a bioprintable composite hydrogel with similarities to the microscopic architecture of a native tumor stroma. We studied the printability of the composite hydrogel via rheology and obtained the optimal printing window. Breast cancer cells and fibroblasts were embedded in the hydrogels and printed to form a 3D model mimicking the in vivo microenvironment. The bioprinted heterogeneous model achieves a high viability for long-term cell culture (> 30 days) and promotes the self-assembly of breast cancer cells into multicellular tumor spheroids (MCTS). We observed the migration and interaction of the cancer-associated fibroblast cells (CAFs) with the MCTS in this model. By using bioprinted cell culture platforms as co-culture systems, it offers a unique tool to study the dependence of tumorigenesis on the stroma composition. This technique features a high-throughput, low cost, and high reproducibility, and it can also provide an alternative model to conventional cell monolayer cultures and animal tumor models to study cancer biology.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Mechanical Engineering, McGill University Montreal
| | - Jose Munguia-Lopez
- Department of Bioengineering, McGill University Montreal; Department of Molecular Biology, Instituto Potosino de Investigación Científica y Tecnológica, A.C. (IPICyT)
| | | | - Joel Grant
- Department of Mining and Materials Engineering, McGill University Montreal
| | | | - Antonio De Leon-Rodriguez
- Department of Molecular Biology, Instituto Potosino de Investigación Científica y Tecnológica, A.C. (IPICyT)
| | - Joseph M Kinsella
- Department of Bioengineering, McGill University Montreal; Department of Biomedical Engineering, McGill University Montreal;
| |
Collapse
|
1022
|
Blache U, Vallmajo-Martin Q, Horton ER, Guerrero J, Djonov V, Scherberich A, Erler JT, Martin I, Snedeker JG, Milleret V, Ehrbar M. Notch-inducing hydrogels reveal a perivascular switch of mesenchymal stem cell fate. EMBO Rep 2018; 19:embr.201845964. [PMID: 29967223 DOI: 10.15252/embr.201845964] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/01/2018] [Accepted: 06/08/2018] [Indexed: 12/26/2022] Open
Abstract
The fate of mesenchymal stem cells (MSCs) in the perivascular niche, as well as factors controlling their fate, is poorly understood. Here, we study MSCs in the perivascular microenvironment of endothelial capillaries by modifying a synthetic 3D biomimetic poly(ethylene glycol) (PEG)-hydrogel system in vitro We show that MSCs together with endothelial cells form micro-capillary networks specifically in soft PEG hydrogels. Transcriptome analysis of human MSCs isolated from engineered capillaries shows a prominent switch in extracellular matrix (ECM) production. We demonstrate that the ECM phenotypic switch of MSCs can be recapitulated in the absence of endothelial cells by functionalizing PEG hydrogels with the Notch-activator Jagged1. Moreover, transient culture of MSCs in Notch-inducing microenvironments reveals the reversibility of this ECM switch. These findings provide insight into the perivascular commitment of MSCs by use of engineered niche-mimicking synthetic hydrogels.
Collapse
Affiliation(s)
- Ulrich Blache
- Department of Obstetrics, University Hospital of Zurich, Zurich, Switzerland.,Institute for Biomechanics, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Queralt Vallmajo-Martin
- Department of Obstetrics, University Hospital of Zurich, Zurich, Switzerland.,Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Edward R Horton
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Julien Guerrero
- Department of Biomedicine and Department of Surgery, University Hospital Basel, Basel, Switzerland
| | | | - Arnaud Scherberich
- Department of Biomedicine and Department of Surgery, University Hospital Basel, Basel, Switzerland
| | - Janine T Erler
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Ivan Martin
- Department of Biomedicine and Department of Surgery, University Hospital Basel, Basel, Switzerland
| | - Jess G Snedeker
- Institute for Biomechanics, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland.,Biomechanics Laboratory, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Vincent Milleret
- Department of Obstetrics, University Hospital of Zurich, Zurich, Switzerland
| | - Martin Ehrbar
- Department of Obstetrics, University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
1023
|
Ren X, Yang Q, Yang D, Liang Y, Dong J, Ren Y, Lu X, Xue L, Li L, Xu L. High-strength double network hydrogels as potential materials for artificial 3D scaffold of cell migration in vitro. Colloids Surf A Physicochem Eng Asp 2018. [DOI: 10.1016/j.colsurfa.2018.03.060] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
1024
|
Blache U, Ehrbar M. Inspired by Nature: Hydrogels as Versatile Tools for Vascular Engineering. Adv Wound Care (New Rochelle) 2018; 7:232-246. [PMID: 29984113 PMCID: PMC6032659 DOI: 10.1089/wound.2017.0760] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 10/22/2017] [Indexed: 12/21/2022] Open
Abstract
Significance: Diseases related to vascular malfunction, hyper-vascularization, or lack of vascularization are among the leading causes of morbidity and mortality. Engineered, vascularized tissues as well as angiogenic growth factor-releasing hydrogels could replace defective tissues. Further, treatments and testing of novel vascular therapeutics will benefit significantly from models that allow for the study of vascularized tissues under physiological relevant in vitro conditions. Recent Advances: Inspired by fibrin, the provisional matrix during wound healing, naturally derived and synthetic hydrogel scaffolds have been developed for vascular engineering. Today, engineers and biologists use commercially available hydrogels to pre-vascularize tissues, to control the delivery of angiogenic growth factors, and to establish vascular diseases models. Critical Issue: For clinical translation, pre-vascularized tissue constructs must be sufficiently large and stable to substitute function-relevant tissue defects and integrate with host vascular perfusion. Moreover, the continuous integration of knowhow from basic vascular biology with innovative, tailorable materials and advanced manufacturing technologies is key to achieving near-physiological tissue models and new treatments to control vascularization. Future Directions: For transplantation, engineered tissues must comprise hierarchically organized vascular trees of different caliber and function. The development of novel vascularization-promoting or -inhibiting therapeutics will benefit from physiologically relevant vessel models. In addition, tissue models representing treatment-relevant vascular tissue functions will increase the capacity to screen for therapeutic compounds and will significantly reduce the need for animals for their validation.
Collapse
Affiliation(s)
- Ulrich Blache
- Department of Obstetrics, University and University Hospital Zurich, Zurich, Switzerland
- Department of Health Sciences and Technology, Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Martin Ehrbar
- Department of Obstetrics, University and University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
1025
|
Sun G, Shen YI, Harmon JW. Engineering Pro-Regenerative Hydrogels for Scarless Wound Healing. Adv Healthc Mater 2018; 7:e1800016. [PMID: 29663707 DOI: 10.1002/adhm.201800016] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 02/17/2018] [Indexed: 12/21/2022]
Abstract
Skin and skin appendages protect the body from harmful environment and prevent internal organs from dehydration. Superficial epidermal wounds usually heal without scarring, however, deep dermal wound healing commonly ends up with nonfunctioning scar formation with substantial loss of skin appendage. Wound healing is one of the most complex dynamic biological processes, during which a cascade of biomolecules combine with stem cell influx and matrix synthesis and synergistically contribute to wound healing at all levels. Although many approaches have been investigated to restore complete skin, the clinically effective therapy is still unavailable and the regeneration of perfect skin still remains a significant challenge. The complete mechanism behind scarless skin regeneration still requires further investigation. Fortunately, recent advancement in regenerative medicine empowers us more than ever to restore tissue in a regenerative manner. Many studies have elucidated and reviewed the contribution of stem cells and growth factors to scarless wound healing. This article focuses on recent advances in scarless wound healing, especially strategies to engineer pro-regenerative scaffolds to restore damaged skin in a regenerative manner.
Collapse
Affiliation(s)
- Guoming Sun
- Sunogel Biotechnologies Inc.; 9 W Ridgely Road Ste 270 Lutherville Timonium MD 21093 USA
| | - Yu-I Shen
- Sunogel Biotechnologies Inc.; 9 W Ridgely Road Ste 270 Lutherville Timonium MD 21093 USA
| | - John W. Harmon
- Department of Surgery and the Hendrix Burn Lab; Johns Hopkins University School of Medicine; Baltimore MD 21224 USA
| |
Collapse
|
1026
|
RNA extraction from self-assembling peptide hydrogels to allow qPCR analysis of encapsulated cells. PLoS One 2018; 13:e0197517. [PMID: 29864116 PMCID: PMC5986125 DOI: 10.1371/journal.pone.0197517] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 05/03/2018] [Indexed: 12/21/2022] Open
Abstract
Self-assembling peptide hydrogels offer a novel 3-dimensional platform for many applications in cell culture and tissue engineering but are not compatible with current methods of RNA isolation; owing to interactions between RNA and the biomaterial. This study investigates the use of two techniques based on two different basic extraction principles: solution-based extraction and direct solid-state binding of RNA respectively, to extract RNA from cells encapsulated in four β-sheet forming self-assembling peptide hydrogels with varying net positive charge. RNA-peptide fibril interactions, rather than RNA-peptide molecular complexing, were found to interfere with the extraction process resulting in low yields. A column-based approach relying on RNA-specific binding was shown to be more suited to extracting RNA with higher purity from these peptide hydrogels owing to its reliance on strong specific RNA binding interactions which compete directly with RNA-peptide fibril interactions. In order to reduce the amount of fibrils present and improve RNA yields a broad spectrum enzyme solution-pronase-was used to partially digest the hydrogels before RNA extraction. This pre-treatment was shown to significantly increase the yield of RNA extracted, allowing downstream RT-qPCR to be performed.
Collapse
|
1027
|
Collagen/chitosan/hyaluronic acid - based injectable hydrogels for tissue engineering applications - design, physicochemical and biological characterization. Colloids Surf B Biointerfaces 2018; 170:152-162. [PMID: 29902729 DOI: 10.1016/j.colsurfb.2018.06.004] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 05/20/2018] [Accepted: 06/04/2018] [Indexed: 12/22/2022]
Abstract
Studies on synthesis, physico-chemical and biological properties of novel biomimetic materials, potentially useful as injectable hydrogels are presented. These materials are in situ prepared chemically crosslinked collagen/chitosan/hyaluronic acid-based hydrogels exhibiting potential for tissue regeneration. Optimization of hydrogels involved testing the effect of various concentration of crosslinking agent (genipin) as well as different ratios of biopolymers used on their properties. The changes in the content of hyaluronic acid and in the genipin concentration used have been shown to be crucial. Employing the highest concentration of crosslinking agent studied (20 mM) the hydrogels of compact structure, characterized by good mechanical properties and prolonged degradation profile can be obtained. Changing the HA content in sol mixture the hydrogel of various wettability; more or less hydrophilic when compared to pure collagen/chitosan hydrogels can be fabricated. The in vitro cell culture study has shown that the surface of the prepared materials ensures suitable biocompatibility. These hydrogels can support the proliferation and adhesion of MG-63 cell line as it was demonstrated using Alamar Blue assay and SEM observations. It is believed that the collagen/chitosan/hyaluronic acid hydrogels crosslinked with genipin are particularly promising materials for bone regeneration procedures, especially attractive for regeneration of small bone losses. This is the first paper in the litearature presenting results of studies on that type of biopolymeric injectable hydrogels chemically crosslinked with genipin.
Collapse
|
1028
|
Kontturi E, Laaksonen P, Linder MB, Gröschel AH, Rojas OJ, Ikkala O. Advanced Materials through Assembly of Nanocelluloses. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1703779. [PMID: 29504161 DOI: 10.1002/adma.201703779] [Citation(s) in RCA: 340] [Impact Index Per Article: 56.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/06/2017] [Indexed: 05/20/2023]
Abstract
There is an emerging quest for lightweight materials with excellent mechanical properties and economic production, while still being sustainable and functionalizable. They could form the basis of the future bioeconomy for energy and material efficiency. Cellulose has long been recognized as an abundant polymer. Modified celluloses were, in fact, among the first polymers used in technical applications; however, they were later replaced by petroleum-based synthetic polymers. Currently, there is a resurgence of interest to utilize renewable resources, where cellulose is foreseen to make again a major impact, this time in the development of advanced materials. This is because of its availability and properties, as well as economic and sustainable production. Among cellulose-based structures, cellulose nanofibrils and nanocrystals display nanoscale lateral dimensions and lengths ranging from nanometers to micrometers. Their excellent mechanical properties are, in part, due to their crystalline assembly via hydrogen bonds. Owing to their abundant surface hydroxyl groups, they can be easily modified with nanoparticles, (bio)polymers, inorganics, or nanocarbons to form functional fibers, films, bulk matter, and porous aerogels and foams. Here, some of the recent progress in the development of advanced materials within this rapidly growing field is reviewed.
Collapse
Affiliation(s)
- Eero Kontturi
- Department of Bioproducts and Biosystems, Aalto University, Espoo, FI-00076, Finland
| | - Päivi Laaksonen
- Department of Bioproducts and Biosystems, Aalto University, Espoo, FI-00076, Finland
- Center of Excellence Molecular Engineering of Biosynthetic Hybrid Materials Research, Aalto University and VTT, Espoo, FI-00076, Finland
| | - Markus B Linder
- Department of Bioproducts and Biosystems, Aalto University, Espoo, FI-00076, Finland
- Center of Excellence Molecular Engineering of Biosynthetic Hybrid Materials Research, Aalto University and VTT, Espoo, FI-00076, Finland
| | - André H Gröschel
- Physical Chemistry and Centre for Nanointegration (CENIDE), University of Duisburg-Essen, DE-45127, Essen, Germany
| | - Orlando J Rojas
- Department of Bioproducts and Biosystems, Aalto University, Espoo, FI-00076, Finland
- Center of Excellence Molecular Engineering of Biosynthetic Hybrid Materials Research, Aalto University and VTT, Espoo, FI-00076, Finland
- Department of Applied Physics, Aalto University, Espoo, FI-00076, Finland
| | - Olli Ikkala
- Department of Bioproducts and Biosystems, Aalto University, Espoo, FI-00076, Finland
- Center of Excellence Molecular Engineering of Biosynthetic Hybrid Materials Research, Aalto University and VTT, Espoo, FI-00076, Finland
- Department of Applied Physics, Aalto University, Espoo, FI-00076, Finland
| |
Collapse
|
1029
|
Centeno EGZ, Cimarosti H, Bithell A. 2D versus 3D human induced pluripotent stem cell-derived cultures for neurodegenerative disease modelling. Mol Neurodegener 2018; 13:27. [PMID: 29788997 PMCID: PMC5964712 DOI: 10.1186/s13024-018-0258-4] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 05/08/2018] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD) and amyotrophic lateral sclerosis (ALS), affect millions of people every year and so far, there are no therapeutic cures available. Even though animal and histological models have been of great aid in understanding disease mechanisms and identifying possible therapeutic strategies, in order to find disease-modifying solutions there is still a critical need for systems that can provide more predictive and physiologically relevant results. One possible avenue is the development of patient-derived models, e.g. by reprogramming patient somatic cells into human induced pluripotent stem cells (hiPSCs), which can then be differentiated into any cell type for modelling. These systems contain key genetic information from the donors, and therefore have enormous potential as tools in the investigation of pathological mechanisms underlying disease phenotype, and progression, as well as in drug testing platforms. hiPSCs have been widely cultured in 2D systems, but in order to mimic human brain complexity, 3D models have been proposed as a more advanced alternative. This review will focus on the use of patient-derived hiPSCs to model AD, PD, HD and ALS. In brief, we will cover the available stem cells, types of 2D and 3D culture systems, existing models for neurodegenerative diseases, obstacles to model these diseases in vitro, and current perspectives in the field.
Collapse
Affiliation(s)
- Eduarda G Z Centeno
- Department of Biotechnology, Federal University of Pelotas, Campus Capão do Leão, Pelotas, RS, 96160-000, Brazil.,Department of Pharmacology, Federal University of Santa Catarina, Campus Trindade, Florianópolis, SC, 88040-900, Brazil
| | - Helena Cimarosti
- Department of Pharmacology, Federal University of Santa Catarina, Campus Trindade, Florianópolis, SC, 88040-900, Brazil.
| | - Angela Bithell
- School of Pharmacy, University of Reading, Whiteknights Campus, Reading, RG6 6UB, UK.
| |
Collapse
|
1030
|
Aeby EA, Misun PM, Hierlemann A, Frey O. Microfluidic Hydrogel Hanging-Drop Network for Long-Term Culturing of 3D Microtissues and Simultaneous High-Resolution Imaging. ACTA ACUST UNITED AC 2018. [DOI: 10.1002/adbi.201800054] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Elise A. Aeby
- Bio Engineering Laboratory; Department of Biosystems Science and Engineering; ETH Zürich; Mattenstrasse 26 4058 Basel Switzerland
| | - Patrick M. Misun
- Bio Engineering Laboratory; Department of Biosystems Science and Engineering; ETH Zürich; Mattenstrasse 26 4058 Basel Switzerland
| | - Andreas Hierlemann
- Bio Engineering Laboratory; Department of Biosystems Science and Engineering; ETH Zürich; Mattenstrasse 26 4058 Basel Switzerland
| | - Olivier Frey
- Bio Engineering Laboratory; Department of Biosystems Science and Engineering; ETH Zürich; Mattenstrasse 26 4058 Basel Switzerland
| |
Collapse
|
1031
|
Kanda P, Alarcon EI, Yeuchyk T, Parent S, de Kemp RA, Variola F, Courtman D, Stewart DJ, Davis DR. Deterministic Encapsulation of Human Cardiac Stem Cells in Variable Composition Nanoporous Gel Cocoons To Enhance Therapeutic Repair of Injured Myocardium. ACS NANO 2018; 12:4338-4350. [PMID: 29660269 DOI: 10.1021/acsnano.7b08881] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Although cocooning explant-derived cardiac stem cells (EDCs) in protective nanoporous gels (NPGs) prior to intramyocardial injection boosts long-term cell retention, the number of EDCs that finally engraft is trivial and unlikely to account for salutary effects on myocardial function and scar size. As such, we investigated the effect of varying the NPG content within capsules to alter the physical properties of cocoons without influencing cocoon dimensions. Increasing NPG concentration enhanced cell migration and viability while improving cell-mediated repair of injured myocardium. Given that the latter occurred with NPG content having no detectable effect on the long-term engraftment of transplanted cells, we found that changing the physical properties of cocoons prompted explant-derived cardiac stem cells to produce greater amounts of cytokines, nanovesicles, and microRNAs that boosted the generation of new blood vessels and new cardiomyocytes. Thus, by altering the physical properties of cocoons by varying NPG content, the paracrine signature of encapsulated cells can be enhanced to promote greater endogenous repair of injured myocardium.
Collapse
Affiliation(s)
- Pushpinder Kanda
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine , University of Ottawa , Ottawa , Canada K1Y4W7
| | - Emilio I Alarcon
- Division of Cardiac Surgery, Department of Surgery, University of Ottawa Heart Institute , University of Ottawa , Ottawa , Canada K1Y4W7
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine , University of Ottawa , Ottawa , Canada K1H8M5
| | - Tanya Yeuchyk
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine , University of Ottawa , Ottawa , Canada K1Y4W7
| | - Sandrine Parent
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine , University of Ottawa , Ottawa , Canada K1Y4W7
| | - Robert A de Kemp
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine , University of Ottawa , Ottawa , Canada K1Y4W7
| | - Fabio Variola
- Department of Mechanical Engineering , University of Ottawa , Ottawa , Canada K1N6N5
- Department of Cellular and Molecular Medicine , University of Ottawa , Ottawa , Canada K1H8M5
| | - David Courtman
- Regenerative Medicine Program , Ottawa Hospital Research Institute , Ottawa , Canada K1H8L6
| | - Duncan J Stewart
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine , University of Ottawa , Ottawa , Canada K1Y4W7
- Department of Cellular and Molecular Medicine , University of Ottawa , Ottawa , Canada K1H8M5
- Regenerative Medicine Program , Ottawa Hospital Research Institute , Ottawa , Canada K1H8L6
| | - Darryl R Davis
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine , University of Ottawa , Ottawa , Canada K1Y4W7
- Department of Cellular and Molecular Medicine , University of Ottawa , Ottawa , Canada K1H8M5
| |
Collapse
|
1032
|
Abstract
Self-assembled peptide nanostructures have been increasingly exploited as functional materials for applications in biomedicine and energy. The emergent properties of these nanomaterials determine the applications for which they can be exploited. It has recently been appreciated that nanomaterials composed of multicomponent coassembled peptides often display unique emergent properties that have the potential to dramatically expand the functional utility of peptide-based materials. This review presents recent efforts in the development of multicomponent peptide assemblies. The discussion includes multicomponent assemblies derived from short low molecular weight peptides, peptide amphiphiles, coiled coil peptides, collagen, and β-sheet peptides. The design, structure, emergent properties, and applications for these multicomponent assemblies are presented in order to illustrate the potential of these formulations as sophisticated next-generation bio-inspired materials.
Collapse
Affiliation(s)
- Danielle M Raymond
- Department of Chemistry, University of Rochester, Rochester, NY 14627-0216, USA.
| | | |
Collapse
|
1033
|
Abstract
Three-dimensional (3D) cell culture systems have gained increasing interest not only for 3D migration studies but also for their use in drug screening, tissue engineering, and ex vivo modeling of metastatic behavior in the field of cancer biology and morphogenesis in the field of developmental biology. The goal of studying cells in a 3D context is to attempt to more faithfully recapitulate the physiological microenvironment of tissues, including mechanical and structural parameters that we envision will reveal more predictive data for development programs and disease states. In this review, we discuss the pros and cons of several well-characterized 3D cell culture systems for performing 3D migration studies. We discuss the intracellular and extracellular signaling mechanisms that govern cell migration. We also describe the mathematical models and relevant assumptions that can be used to describe 3D cell movement.
Collapse
Affiliation(s)
- Pei-Hsun Wu
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences in Oncology Center, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, USA;, ,
| | - Daniele M. Gilkes
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences in Oncology Center, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, USA;, ,
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Denis Wirtz
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences in Oncology Center, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, USA;, ,
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
1034
|
LeSavage BL, Suhar NA, Madl CM, Heilshorn SC. Production of Elastin-like Protein Hydrogels for Encapsulation and Immunostaining of Cells in 3D. J Vis Exp 2018. [PMID: 29863669 DOI: 10.3791/57739] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Two-dimensional (2D) tissue culture techniques have been essential for our understanding of fundamental cell biology. However, traditional 2D tissue culture systems lack a three-dimensional (3D) matrix, resulting in a significant disconnect between results collected in vitro and in vivo. To address this limitation, researchers have engineered 3D hydrogel tissue culture platforms that can mimic the biochemical and biophysical properties of the in vivo cell microenvironment. This research has motivated the need to develop material platforms that support 3D cell encapsulation and downstream biochemical assays. Recombinant protein engineering offers a unique toolset for 3D hydrogel material design and development by allowing for the specific control of protein sequence and therefore, by extension, the potential mechanical and biochemical properties of the resultant matrix. Here, we present a protocol for the expression of recombinantly-derived elastin-like protein (ELP), which can be used to form hydrogels with independently tunable mechanical properties and cell-adhesive ligand concentration. We further present a methodology for cell encapsulation within ELP hydrogels and subsequent immunofluorescent staining of embedded cells for downstream analysis and quantification.
Collapse
Affiliation(s)
| | - Nicholas A Suhar
- Department of Materials Science and Engineering, Stanford University
| | | | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University;
| |
Collapse
|
1035
|
Mukhopadhyay RD, Das G, Ajayaghosh A. Stepwise control of host-guest interaction using a coordination polymer gel. Nat Commun 2018; 9:1987. [PMID: 29777098 PMCID: PMC5959896 DOI: 10.1038/s41467-018-04303-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 04/10/2018] [Indexed: 11/09/2022] Open
Abstract
Precise control of host-guest interaction as seen in biological processes is difficult to achieve with artificial systems. Herein we have exploited the thermodynamic benefits of a system in equilibrium to achieve controlled stepwise release and capture of cyclodextrin (guest) using a coordination polymer (Mg-CP) as the host and temperature as the stimulus. Since temperature is not a precision stimulus for artificial host-guest interaction, the present system is a distinct prototype that manifests temperature-controlled natural host-guest interaction. The described coordination polymeric host system, when incorporated into a hydrogel matrix, provides a microenvironment that facilitates the stepwise release of α-CD in response to temperature variation within a quasi-solid state. The work demonstrated here may pave the way towards thermally controlled delivery and monitoring of otherwise spectroscopically silent molecules such as cyclodextrins.
Collapse
Affiliation(s)
- Rahul Dev Mukhopadhyay
- Photosciences and Photonics Section, Chemical Sciences and Technology Division and Academy of Scientific and Innovative Research (AcSIR), CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram, 695019, India
| | - Gourab Das
- Photosciences and Photonics Section, Chemical Sciences and Technology Division and Academy of Scientific and Innovative Research (AcSIR), CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram, 695019, India
| | - Ayyappanpillai Ajayaghosh
- Photosciences and Photonics Section, Chemical Sciences and Technology Division and Academy of Scientific and Innovative Research (AcSIR), CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram, 695019, India.
| |
Collapse
|
1036
|
Kabb CP, O'Bryan CS, Deng CC, Angelini TE, Sumerlin BS. Photoreversible Covalent Hydrogels for Soft-Matter Additive Manufacturing. ACS APPLIED MATERIALS & INTERFACES 2018; 10:16793-16801. [PMID: 29726251 DOI: 10.1021/acsami.8b02441] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Reversible covalent chemistry provides access to robust materials with the ability to be degraded and reformed upon exposure to an appropriate stimulus. Photoresponsive units are attractive for this purpose, as the spatial and temporal application of light is easily controlled. Coumarin derivatives undergo a [2 + 2] cycloaddition upon exposure to long-wave UV irradiation (365 nm), and this process can be reversed using short-wave UV light (254 nm). Therefore, polymers cross-linked by coumarin groups are excellent candidates as reversible covalent gels. In this work, copolymerization of coumarin-containing monomers with the hydrophilic comonomer N, N-dimethylacrylamide yielded water-soluble, linear polymers that could be cured with long-wave UV light into free-standing hydrogels, even in the absence of a photoinitiator. Importantly, the gels were reverted back to soluble copolymers upon short-wave UV irradiation. This process could be cycled, allowing for recycling and remolding of the hydrogel into additional shapes. Further, this hydrogel can be imprinted with patterns through a mask-based, post-gelation photoetching method. Traditional limitations of this technique, such as the requirement for uniform etching in one direction, have been overcome by combining these materials with a soft-matter additive manufacturing methodology. In a representative application of this approach, we printed solid structures in which the interior coumarin-cross-linked gel is surrounded by a nondegradable gel. Upon exposure to short-wave UV irradiation, the coumarin-cross-linked gel was reverted to soluble prepolymers that were washed away to yield hollow hydrogel objects.
Collapse
|
1037
|
Mineral Surface-Templated Self-Assembling Systems: Case Studies from Nanoscience and Surface Science towards Origins of Life Research. Life (Basel) 2018; 8:life8020010. [PMID: 29738443 PMCID: PMC6027067 DOI: 10.3390/life8020010] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 04/26/2018] [Accepted: 05/03/2018] [Indexed: 01/20/2023] Open
Abstract
An increasing body of evidence relates the wide range of benefits mineral surfaces offer for the development of early living systems, including adsorption of small molecules from the aqueous phase, formation of monomeric subunits and their subsequent polymerization, and supramolecular assembly of biopolymers and other biomolecules. Each of these processes was likely a necessary stage in the emergence of life on Earth. Here, we compile evidence that templating and enhancement of prebiotically-relevant self-assembling systems by mineral surfaces offers a route to increased structural, functional, and/or chemical complexity. This increase in complexity could have been achieved by early living systems before the advent of evolvable systems and would not have required the generally energetically unfavorable formation of covalent bonds such as phosphodiester or peptide bonds. In this review we will focus on various case studies of prebiotically-relevant mineral-templated self-assembling systems, including supramolecular assemblies of peptides and nucleic acids, from nanoscience and surface science. These fields contain valuable information that is not yet fully being utilized by the origins of life and astrobiology research communities. Some of the self-assemblies that we present can promote the formation of new mineral surfaces, similar to biomineralization, which can then catalyze more essential prebiotic reactions; this could have resulted in a symbiotic feedback loop by which geology and primitive pre-living systems were closely linked to one another even before life’s origin. We hope that the ideas presented herein will seed some interesting discussions and new collaborations between nanoscience/surface science researchers and origins of life/astrobiology researchers.
Collapse
|
1038
|
Peng X, Peng Y, Han B, Liu W, Zhang F, Linhardt RJ. IO4−-stimulated crosslinking of catechol-conjugated hydroxyethyl chitosan as a tissue adhesive. J Biomed Mater Res B Appl Biomater 2018; 107:582-593. [DOI: 10.1002/jbm.b.34150] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 02/26/2018] [Accepted: 04/17/2018] [Indexed: 12/21/2022]
Affiliation(s)
- Xiaoting Peng
- College of Marine Life Sciences, Ocean University of China; Qingdao China
| | - Yanfei Peng
- College of Marine Life Sciences, Ocean University of China; Qingdao China
| | - Baoqin Han
- College of Marine Life Sciences, Ocean University of China; Qingdao China
| | - Wanshun Liu
- College of Marine Life Sciences, Ocean University of China; Qingdao China
| | - Fuming Zhang
- Department of Chemistry and Chemical Biology; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute; Troy New York 12180
- Department of Chemical and Biological Engineering; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute; Troy New York 12180
- Department of Biology; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute; Troy New York 12180
- Department of Biomedical Engineering; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute; Troy New York 12180
| | - Robert J Linhardt
- Department of Chemistry and Chemical Biology; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute; Troy New York 12180
- Department of Chemical and Biological Engineering; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute; Troy New York 12180
- Department of Biology; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute; Troy New York 12180
- Department of Biomedical Engineering; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute; Troy New York 12180
| |
Collapse
|
1039
|
Axpe E, Duraj-Thatte A, Chang Y, Kaimaki DM, Sanchez-Sanchez A, Caliskan HB, Dorval Courchesne NM, Joshi NS. Fabrication of Amyloid Curli Fibers–Alginate Nanocomposite Hydrogels with Enhanced Stiffness. ACS Biomater Sci Eng 2018; 4:2100-2105. [DOI: 10.1021/acsbiomaterials.8b00364] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Eneko Axpe
- Nanoscience Centre, Department of Engineering, Cambridge University, 11 JJ Thomson Ave., Cambridge CB3 0FF, United Kingdom
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Cir., Boston, Massachusetts 02115, United States
- School of Engineering and Applied Sciences, Harvard University, 29 Oxford St., Cambridge, Massachusetts 02138, United States
| | - Anna Duraj-Thatte
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Cir., Boston, Massachusetts 02115, United States
- School of Engineering and Applied Sciences, Harvard University, 29 Oxford St., Cambridge, Massachusetts 02138, United States
| | - Yin Chang
- Nanoscience Centre, Department of Engineering, Cambridge University, 11 JJ Thomson Ave., Cambridge CB3 0FF, United Kingdom
| | - Domna-Maria Kaimaki
- Nanoscience Centre, Department of Engineering, Cambridge University, 11 JJ Thomson Ave., Cambridge CB3 0FF, United Kingdom
| | - Ana Sanchez-Sanchez
- Nanoscience Centre, Department of Engineering, Cambridge University, 11 JJ Thomson Ave., Cambridge CB3 0FF, United Kingdom
- Electrical Engineering Division, Department of Engineering, Cambridge University, Trumpington St., Cambridge CB2 1PZ, United Kingdom
| | - H. Burak Caliskan
- Nanoscience Centre, Department of Engineering, Cambridge University, 11 JJ Thomson Ave., Cambridge CB3 0FF, United Kingdom
| | - Noémie-Manuelle Dorval Courchesne
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Cir., Boston, Massachusetts 02115, United States
- School of Engineering and Applied Sciences, Harvard University, 29 Oxford St., Cambridge, Massachusetts 02138, United States
| | - Neel S. Joshi
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Cir., Boston, Massachusetts 02115, United States
- School of Engineering and Applied Sciences, Harvard University, 29 Oxford St., Cambridge, Massachusetts 02138, United States
| |
Collapse
|
1040
|
Thomas AK, Wieduwild R, Zimmermann R, Lin W, Friedrichs J, Bickle M, Fahmy K, Werner C, Zhang Y. Layer-by-Layer Assembly of Heparin and Peptide-Polyethylene Glycol Conjugates to Form Hybrid Nanothin Films of Biomatrices. ACS APPLIED MATERIALS & INTERFACES 2018; 10:14264-14270. [PMID: 29658265 DOI: 10.1021/acsami.8b02014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
We investigated the utility of a heparin/peptide-polyethylene glycol conjugate system to build layer-by-layer (LbL) structures, to assemble tailored multilayer-biomatrices for cell culture. The LbL assembly balances the advantages of polyelectrolyte systems and protein-based systems. Human umbilical vein endothelial cells showed distinct responses to the film thickness and structure; the presence, density, and spatial arrangement of a cell adhesion ligand within the nanothin film; and the pretreatment of the film with morphogens. The LbL technique presents a versatile tool for modifying cell culture substrates with defined and diverse biochemical and structural features, for investigating cell-material interactions.
Collapse
Affiliation(s)
| | | | - Ralf Zimmermann
- Department of Biofunctional Polymer Materials, Max Bergman Center of Biomaterials , Leibniz Institute of Polymer Research , Dresden 01069 , Germany
| | | | - Jens Friedrichs
- Department of Biofunctional Polymer Materials, Max Bergman Center of Biomaterials , Leibniz Institute of Polymer Research , Dresden 01069 , Germany
| | - Marc Bickle
- High-Throughput Technology Development Studio , Max Planck Institute of Molecular Cell Biology and Genetics , Dresden 01307 , Germany
| | | | - Carsten Werner
- Department of Biofunctional Polymer Materials, Max Bergman Center of Biomaterials , Leibniz Institute of Polymer Research , Dresden 01069 , Germany
| | | |
Collapse
|
1041
|
Bello AB, Park H, Lee SH. Current approaches in biomaterial-based hematopoietic stem cell niches. Acta Biomater 2018; 72:1-15. [PMID: 29578087 DOI: 10.1016/j.actbio.2018.03.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 02/07/2018] [Accepted: 03/14/2018] [Indexed: 12/20/2022]
Abstract
Hematopoietic stem cells (HSCs) are multipotent progenitor cells that can differentiate and replenish blood and immune cells. While there is a growing demand for autologous and allogeneic HSC transplantation owing to the increasing incidence of hereditary and hematologic diseases, the low population of HSCs in cord-blood and bone marrow (the main source of HSCs) hinders their medical applicability. Several cytokine and growth factor-based methods have been developed to expand the HSCs in vitro; however, the expansion rate is low, or the expanded cells fail to survive upon engraftment. This is at least in part because the overly simplistic polystyrene culture substrates fail to fully replicate the microenvironments or niches where these stem cells live. Bone marrow niches are multi-dimensional, complex systems that involve both biochemical (cells, growth factors, and cytokines) and physiochemical (stiffness, O2 concentration, and extracellular matrix presentation) factors that regulate the quiescence, proliferation, activation, and differentiation of the HSCs. Although several studies have been conducted on in vitro HSC expansion via 2D and 3D biomaterial-based platforms, additional work is required to engineer an effective biomaterial platform that mimics bone marrow niches. In this study, the factors that regulate the HSC in vivo were explained and their applications in the engineering of a bone marrow biomaterial-based platform were discussed. In addition, current approaches, challenges, and the future direction of a biomaterial-based culture and expansion of the HSC were examined. STATEMENT OF SIGNIFICANCE Hematopoietic stem cells (HSC) are multipotent cells that can differentiate and replace the blood and immune cells of the body. However, in vivo, there is a low population of these cells, and thus their use in biotherapeutic and medical applications is limited (i.e., bone marrow transplantation). In this review, the biochemical factors (growth factors, cytokines, co-existing cells, ECM, gas concentrations, and differential gene expression) that may regulate the over-all fate of HSC, in vivo, were summarized and discussed. Moreover, different conventional and recent biomaterial platforms were reviewed, and their potential in generating a biomaterial-based, BM niche-mimicking platform for the efficient growth and expansion of clinically relevant HSCs in-vitro, was discussed.
Collapse
Affiliation(s)
- Alvin Bacero Bello
- School of Integrative Engineering, Chung-Ang University, Seoul 06911, Republic of Korea; Department of Biomedical Science, CHA University, Seongnam-Si 13488, Republic of Korea
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul 06911, Republic of Korea.
| | - Soo-Hong Lee
- Department of Biomedical Science, CHA University, Seongnam-Si 13488, Republic of Korea.
| |
Collapse
|
1042
|
González de Torre I, Ibáñez-Fonseca A, Quintanilla L, Alonso M, Rodríguez-Cabello JC. Random and oriented electrospun fibers based on a multicomponent, in situ clickable elastin-like recombinamer system for dermal tissue engineering. Acta Biomater 2018; 72:137-149. [PMID: 29574183 DOI: 10.1016/j.actbio.2018.03.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 02/20/2018] [Accepted: 03/14/2018] [Indexed: 10/17/2022]
Abstract
Herein we present a system to obtain fibers from clickable elastin-like recombinamers (ELRs) that crosslink in situ during the electrospinning process itself, with no need for any further treatment to stabilize them. These ELR-click fibers are completely stable under in vitro conditions. A wrinkled fiber morphology is obtained. In addition to a random fiber orientation, oriented fibers with a high degree of alignment and coherence can also be obtained by using a rotational electrode. The production of multicomponent fibers means that different functionalities, such as cell-adhesion domains (RGD peptides), can be incorporated into them. In a subsequent study, two main cell lines present in the dermis and epidermis, namely keratinocytes and fibroblasts, were cultured on top of the ELR-click fibers. Adhesion, proliferation, fluorescence, immunostaining and histology studies showed the cytocompatibility of these scaffolds, thus suggesting their possible use for wound dressings in skin tissue engineering applications. STATEMENT OF SIGNIFICANCE For the first time stable electrospun bioactive fibers are obtained by the in situ mixing of two "clickable" ELR components previously described by Gonzalez et al (Acta Biomaterialia 2014). This work describes an efficient system to prepare fibrous scaffolds based on peptidic polymers by electrospinning without the need of crosslinking agents that could be harmful for cells or living tissues. These bioactive fibers support cell growth due to the inclusion of RGD motifs (Staubli et al. Biomaterials 2017). Finally, the in vitro biocompatibility of the two main cell types found in the outer layers of skin, fibroblasts and keratinocytes, indicates that this system is of great interest to prepare elastic artificial skin substitutes for wound healing applications.
Collapse
|
1043
|
Li S, Dong S, Xu W, Tu S, Yan L, Zhao C, Ding J, Chen X. Antibacterial Hydrogels. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1700527. [PMID: 29876202 PMCID: PMC5980143 DOI: 10.1002/advs.201700527] [Citation(s) in RCA: 554] [Impact Index Per Article: 92.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 12/29/2017] [Indexed: 05/03/2023]
Abstract
Antibacterial materials are recognized as important biomaterials due to their effective inhibition of bacterial infections. Hydrogels are 3D polymer networks crosslinked by either physical interactions or covalent bonds. Currently, hydrogels with an antibacterial function are a main focus in biomedical research. Many advanced antibacterial hydrogels are developed, each possessing unique qualities, namely high water swellability, high oxygen permeability, improved biocompatibility, ease of loading and releasing drugs, and structural diversity. Here, an overview of the structures, performances, mechanisms of action, loading and release behaviors, and applications of various antibacterial hydrogel formulations is provided. Furthermore, the prospects in biomedical research and clinical applications are predicted.
Collapse
Affiliation(s)
- Shuqiang Li
- Department of Bone and Joint SurgeryThe First Hospital of Jilin UniversityChangchun130022P. R. China
| | - Shujun Dong
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022P. R. China
- VIP Integrated DepartmentSchool and Hospital of Stomatology Jilin UniversityChangchun130021P. R. China
| | - Weiguo Xu
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022P. R. China
| | - Shicheng Tu
- Department of Bone and Joint SurgeryThe First Hospital of Jilin UniversityChangchun130022P. R. China
| | - Lesan Yan
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022P. R. China
| | - Changwen Zhao
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022P. R. China
| |
Collapse
|
1044
|
Burgstaller G, Sengupta A, Vierkotten S, Preissler G, Lindner M, Behr J, Königshoff M, Eickelberg O. Distinct niches within the extracellular matrix dictate fibroblast function in (cell free) 3D lung tissue cultures. Am J Physiol Lung Cell Mol Physiol 2018; 314:L708-L723. [DOI: 10.1152/ajplung.00408.2017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Cues from the extracellular matrix (ECM) and their functional interplay with cells play pivotal roles for development, tissue repair, and disease. However, the precise nature of this interplay remains elusive. We used an innovative 3D cell culture ECM model by decellularizing 300-µm-thick ex vivo lung tissue scaffolds (d3D-LTCs) derived from diseased and healthy mouse lungs, which widely mimics the native (patho)physiological in vivo ECM microenvironment. We successfully repopulated all d3D-LTCs with primary human and murine fibroblasts, and moreover, we demonstrated that the cells also populated the innermost core regions of the d3D-LTCs in a real 3D fashion. The engrafted fibroblasts revealed a striking functional plasticity, depending on their localization in distinct ECM niches of the d3D-LTCs, affecting the cells’ tissue engraftment, cellular migration rates, cell morphologies, and protein expression and phosphorylation levels. Surprisingly, we also observed fibroblasts that were homing to the lung scaffold’s interstitium as well as fibroblasts that were invading fibrotic areas. To date, the functional nature and even the existence of 3D cell matrix adhesions in vivo as well as in 3D culture models is still unclear and controversial. Here, we show that attachment of fibroblasts to the d3D-LTCs evidently occurred via focal adhesions, thus advocating for a relevant functional role in vivo. Furthermore, we found that protein levels of talin, paxillin, and zyxin and phosphorylation levels of paxillin Y118, as well as the migration-relevant small GTPases RhoA, Rac, and CDC42, were significantly reduced compared with their attachment to 2D plastic dishes. In summary, our results strikingly indicate that inherent physical or compositional characteristics of the ECM act as instructive cues altering the functional behavior of engrafted cells. Thus, d3D-LTCs might aid to obtain more realistic data in vitro, with a high relevance for drug discovery and mechanistic studies alike.
Collapse
Affiliation(s)
- Gerald Burgstaller
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Arunima Sengupta
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Sarah Vierkotten
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Gerhard Preissler
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Thoraxchirurgisches Zentrum, Klinik für Allgemeine-, Viszeral-, Transplantations-, Gefäß- und Thoraxchirurgie, Klinikum Großhadern, Ludwig-Maximilians-Universität, Munich, Germany
| | - Michael Lindner
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Asklepios Fachkliniken München-Gauting, Munich, Germany
| | - Jürgen Behr
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Asklepios Fachkliniken München-Gauting, Medizinische Klinik und Poliklinik V, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany
| | - Melanie Königshoff
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Division of Respiratory Sciences and Critical Care Medicine, University of Colorado, Denver, Colorado
| | - Oliver Eickelberg
- Division of Respiratory Sciences and Critical Care Medicine, University of Colorado, Denver, Colorado
| |
Collapse
|
1045
|
Rödling L, Volz EM, Raic A, Brändle K, Franzreb M, Lee-Thedieck C. Magnetic Macroporous Hydrogels as a Novel Approach for Perfused Stem Cell Culture in 3D Scaffolds via Contactless Motion Control. Adv Healthc Mater 2018; 7:e1701403. [PMID: 29349923 DOI: 10.1002/adhm.201701403] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Indexed: 12/14/2022]
Abstract
There is an urgent need for 3D cell culture systems that avoid the oversimplifications and artifacts of conventional culture in 2D. However, 3D culture within the cavities of porous biomaterials or large 3D structures harboring high cell numbers is limited by the needs to nurture cells and to remove growth-limiting metabolites. To overcome the diffusion-limited transport of such soluble factors in 3D culture, mixing can be improved by pumping, stirring or shaking, but this in turn can lead to other problems. Using pumps typically requires custom-made accessories that are not compatible with conventional cell culture disposables, thus interfering with cell production processes. Stirring or shaking allows little control over movement of scaffolds in media. To overcome these limitations, magnetic, macroporous hydrogels that can be moved or positioned within media in conventional cell culture tubes in a contactless manner are presented. The cytocompatibility of the developed biomaterial and the applied magnetic fields are verified for human hematopoietic stem and progenitor cells (HSPCs). The potential of this technique for perfusing 3D cultures is demonstrated in a proof-of-principle study that shows that controlled contactless movement of cell-laden magnetic hydrogels in culture media can mimic the natural influence of differently perfused environments on HSPCs.
Collapse
Affiliation(s)
- Lisa Rödling
- Karlsruhe Institute of Technology (KIT); Institute of Functional Interfaces; Hermann-von-Helmholtz-Platz 1 76344 Eggenstein-Leopoldshafen Germany
| | - Esther Magano Volz
- Karlsruhe Institute of Technology (KIT); Institute of Functional Interfaces; Hermann-von-Helmholtz-Platz 1 76344 Eggenstein-Leopoldshafen Germany
| | - Annamarija Raic
- Karlsruhe Institute of Technology (KIT); Institute of Functional Interfaces; Hermann-von-Helmholtz-Platz 1 76344 Eggenstein-Leopoldshafen Germany
| | - Katharina Brändle
- Karlsruhe Institute of Technology (KIT); Institute of Functional Interfaces; Hermann-von-Helmholtz-Platz 1 76344 Eggenstein-Leopoldshafen Germany
| | - Matthias Franzreb
- Karlsruhe Institute of Technology (KIT); Institute of Functional Interfaces; Hermann-von-Helmholtz-Platz 1 76344 Eggenstein-Leopoldshafen Germany
| | - Cornelia Lee-Thedieck
- Karlsruhe Institute of Technology (KIT); Institute of Functional Interfaces; Hermann-von-Helmholtz-Platz 1 76344 Eggenstein-Leopoldshafen Germany
| |
Collapse
|
1046
|
Park KM, Shin YM, Kim K, Shin H. Tissue Engineering and Regenerative Medicine 2017: A Year in Review. TISSUE ENGINEERING PART B-REVIEWS 2018; 24:327-344. [PMID: 29652594 DOI: 10.1089/ten.teb.2018.0027] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In 2017, a new paradigm change caused by artificial intelligence and big data analysis resulted in innovation in each field of science and technology, and also significantly influenced progress in tissue engineering and regenerative medicine (TERM). TERM has continued to make technological advances based on interdisciplinary approaches and has contributed to the overall field of biomedical technology, including cancer biology, personalized medicine, development biology, and cell-based therapeutics. While researchers are aware that there is still a long way to go until TERM reaches the ultimate goal of patient treatment through clinical translation, the rapid progress in convergence studies led by technological improvements in TERM has been encouraging. In this review, we highlighted the significant advances made in TERM in 2017 (with an overlap of 5 months in 2016). We identified major progress in TERM in a manner similar to previous reviews published in the last few years. In addition, we carefully considered all four previous reviews during the selection process and chose main themes that minimize the duplication of the topics. Therefore, we have identified three areas that have been the focus of most journal publications in the TERM community in 2017: (i) advanced biomaterials and three-dimensional (3D) cell printing, (ii) exosomes as bioactive agents for regenerative medicine, and (iii) 3D culture in regenerative medicine.
Collapse
Affiliation(s)
- Kyung Min Park
- 1 Division of Bioengineering, Incheon National University , Incheon, Republic of Korea
| | - Young Min Shin
- 2 BioMedical Science Institute, Yonsei University College of Medicine , Seoul, Republic of Korea
| | - Kyobum Kim
- 1 Division of Bioengineering, Incheon National University , Incheon, Republic of Korea
| | - Heungsoo Shin
- 3 Department of Bioengineering, Hanyang University , Seoul, Republic of Korea.,4 BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University , Seoul, Republic of Korea
| |
Collapse
|
1047
|
Maier M, Radtke CP, Hubbuch J, Niemeyer CM, Rabe KS. On-Demand Production of Flow-Reactor Cartridges by 3D Printing of Thermostable Enzymes. Angew Chem Int Ed Engl 2018; 57:5539-5543. [PMID: 29466613 DOI: 10.1002/anie.201711072] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/25/2018] [Indexed: 11/06/2022]
Abstract
The compartmentalization of chemical reactions is an essential principle of life that provides a major source of innovation for the development of novel approaches in biocatalysis. To implement spatially controlled biotransformations, rapid manufacturing methods are needed for the production of biocatalysts that can be applied in flow systems. Whereas three-dimensional (3D) printing techniques offer high-throughput manufacturing capability, they are usually not compatible with the delicate nature of enzymes, which call for physiological processing parameters. We herein demonstrate the utility of thermostable enzymes in the generation of biocatalytic agarose-based inks for a simple temperature-controlled 3D printing process. As examples we utilized an esterase and an alcohol dehydrogenase from thermophilic organisms as well as a decarboxylase that was thermostabilized by directed protein evolution. We used the resulting 3D-printed parts for a continuous, two-step sequential biotransformation in a fluidic setup.
Collapse
Affiliation(s)
- Manfred Maier
- Karlsruhe Institute of Technology (KIT), Institute for Biological Interfaces (IBG 1), Herrmann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Carsten P Radtke
- Karlsruhe Institute of Technology (KIT), Institute for Engineering in Life Science, Section IV: Biomolecular Separation Engineering, Fritz-Haber-Weg 2, 76131, Karlsruhe, Germany
| | - Jürgen Hubbuch
- Karlsruhe Institute of Technology (KIT), Institute for Engineering in Life Science, Section IV: Biomolecular Separation Engineering, Fritz-Haber-Weg 2, 76131, Karlsruhe, Germany
| | - Christof M Niemeyer
- Karlsruhe Institute of Technology (KIT), Institute for Biological Interfaces (IBG 1), Herrmann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Kersten S Rabe
- Karlsruhe Institute of Technology (KIT), Institute for Biological Interfaces (IBG 1), Herrmann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
1048
|
Maier M, Radtke CP, Hubbuch J, Niemeyer CM, Rabe KS. Herstellung direkt nutzbarer Durchflussreaktorkartuschen durch 3D-Druck von thermostabilen Enzymen. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201711072] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Manfred Maier
- Karlsruher Institut für Technologie (KIT); Institut für Biologische Grenzflächen (IBG 1); Herrmann-von-Helmholtz-Platz 1 76344 Eggenstein-Leopoldshafen Deutschland
| | - Carsten P. Radtke
- Karlsruher Institut für Technologie (KIT); Institut für Bio- und Lebensmitteltechnik, Teilinstitut IV: Molekulare Aufarbeitung von Bioprodukten; Fritz-Haber-Weg 2 76131 Karlsruhe Deutschland
| | - Jürgen Hubbuch
- Karlsruher Institut für Technologie (KIT); Institut für Bio- und Lebensmitteltechnik, Teilinstitut IV: Molekulare Aufarbeitung von Bioprodukten; Fritz-Haber-Weg 2 76131 Karlsruhe Deutschland
| | - Christof M. Niemeyer
- Karlsruher Institut für Technologie (KIT); Institut für Biologische Grenzflächen (IBG 1); Herrmann-von-Helmholtz-Platz 1 76344 Eggenstein-Leopoldshafen Deutschland
| | - Kersten S. Rabe
- Karlsruher Institut für Technologie (KIT); Institut für Biologische Grenzflächen (IBG 1); Herrmann-von-Helmholtz-Platz 1 76344 Eggenstein-Leopoldshafen Deutschland
| |
Collapse
|
1049
|
Sitsanidis ED, Piras CC, Alexander BD, Siligardi G, Jávorfi T, Hall AJ, Edwards AA. Circular dichroism studies of low molecular weight hydrogelators: The use of SRCD and addressing practical issues. Chirality 2018; 30:708-718. [PMID: 29645307 DOI: 10.1002/chir.22850] [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: 11/27/2017] [Revised: 02/08/2018] [Accepted: 02/14/2018] [Indexed: 11/08/2022]
Abstract
Circular dichroism (CD) spectroscopy has been used extensively for the investigation of the conformation and configuration of chiral molecules, but its use for evaluating the mode of self-assembly in soft materials has been limited. Herein, we report a protocol for the study of such materials by electronic CD spectroscopy using commercial/benchtop instruments and synchrotron radiation (SR) using the B23 beamline available at Diamond Light Source. The use of the B23 beamtime for SRCD was advantageous because of the unique enhanced spatial resolution achieved because of its highly collimated and small beamlight cross section (ca. 250 μm) and higher photon flux in the far UV region (175-250 nm) enhancing the signal-to-noise ratio relative to benchtop CD instruments. A set of low molecular weight (LMW) hydrogelators, comprising two Fmoc-protected enantiomeric monosaccharides and one Fmoc dipeptide (Fmoc-FF), were studied. The research focused on the optimization of sample preparation and handling, which then enabled the characterization of sample conformational homogeneity and thermal stability. CD spectroscopy, in combination with other spectroscopic techniques and microscopy, will allow a better insight into the self-assembly of chiral building blocks into higher order structural architectures.
Collapse
Affiliation(s)
| | - Carmen C Piras
- Medway School of Pharmacy, Universities of Greenwich and Kent at Medway, Kent, UK
| | - Bruce D Alexander
- Department of Pharmaceutical, Chemical and Environmental Sciences, University of Greenwich, Kent, UK
| | | | | | - Andrew J Hall
- Medway School of Pharmacy, Universities of Greenwich and Kent at Medway, Kent, UK
| | - Alison A Edwards
- Medway School of Pharmacy, Universities of Greenwich and Kent at Medway, Kent, UK
| |
Collapse
|
1050
|
Lambert CR, Nijsure D, Huynh V, Wylie RG. Hydrogels with reversible chemical environments for
in vitro
cell culture. Biomed Mater 2018; 13:045002. [DOI: 10.1088/1748-605x/aab45d] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|