1
|
Ahmed E, Mulay P, Ramirez C, Tirado-Mansilla G, Cheong E, Gormley AJ. Mapping Biomaterial Complexity by Machine Learning. Tissue Eng Part A 2024; 30:662-680. [PMID: 39135398 DOI: 10.1089/ten.tea.2024.0067] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024] Open
Abstract
Biomaterials often have subtle properties that ultimately drive their bespoke performance. Given this nuanced structure-function behavior, the standard scientific approach of one experiment at a time or design of experiment methods is largely inefficient for the discovery of complex biomaterials. More recently, high-throughput experimentation coupled with machine learning methods has matured beyond expert users allowing scientists and engineers from diverse backgrounds to access these powerful data science tools. As a result, we now have the opportunity to strategically utilize all available data from high-throughput experiments to train efficacious models and map the structure-function behavior of biomaterials for their discovery. Herein, we discuss this necessary shift to data-driven determination of structure-function properties of biomaterials as we highlight how machine learning is leveraged in identifying physicochemical cues for biomaterials in tissue engineering, gene delivery, drug delivery, protein stabilization, and antifouling materials. We also discuss data-mining approaches that are coupled with machine learning to map biomaterial functions that reduce the load on experimental approaches for faster biomaterial discovery. Ultimately, harnessing the prowess of machine learning will lead to accelerated discovery and development of optimal biomaterial designs.
Collapse
Affiliation(s)
- Eman Ahmed
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Prajakatta Mulay
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Cesar Ramirez
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Gabriela Tirado-Mansilla
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Eugene Cheong
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Adam J Gormley
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| |
Collapse
|
2
|
Vitorino R. Transforming Clinical Research: The Power of High-Throughput Omics Integration. Proteomes 2024; 12:25. [PMID: 39311198 PMCID: PMC11417901 DOI: 10.3390/proteomes12030025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/31/2024] [Accepted: 09/02/2024] [Indexed: 09/26/2024] Open
Abstract
High-throughput omics technologies have dramatically changed biological research, providing unprecedented insights into the complexity of living systems. This review presents a comprehensive examination of the current landscape of high-throughput omics pipelines, covering key technologies, data integration techniques and their diverse applications. It looks at advances in next-generation sequencing, mass spectrometry and microarray platforms and highlights their contribution to data volume and precision. In addition, this review looks at the critical role of bioinformatics tools and statistical methods in managing the large datasets generated by these technologies. By integrating multi-omics data, researchers can gain a holistic understanding of biological systems, leading to the identification of new biomarkers and therapeutic targets, particularly in complex diseases such as cancer. The review also looks at the integration of omics data into electronic health records (EHRs) and the potential for cloud computing and big data analytics to improve data storage, analysis and sharing. Despite significant advances, there are still challenges such as data complexity, technical limitations and ethical issues. Future directions include the development of more sophisticated computational tools and the application of advanced machine learning techniques, which are critical for addressing the complexity and heterogeneity of omics datasets. This review aims to serve as a valuable resource for researchers and practitioners, highlighting the transformative potential of high-throughput omics technologies in advancing personalized medicine and improving clinical outcomes.
Collapse
Affiliation(s)
- Rui Vitorino
- iBiMED, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal;
- Department of Surgery and Physiology, Cardiovascular R&D Centre—UnIC@RISE, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| |
Collapse
|
3
|
Li J, Cai X, Jiang P, Wang H, Zhang S, Sun T, Chen C, Fan K. Co-based Nanozymatic Profiling: Advances Spanning Chemistry, Biomedical, and Environmental Sciences. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307337. [PMID: 37724878 DOI: 10.1002/adma.202307337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/12/2023] [Indexed: 09/21/2023]
Abstract
Nanozymes, next-generation enzyme-mimicking nanomaterials, have entered an era of rational design; among them, Co-based nanozymes have emerged as captivating players over times. Co-based nanozymes have been developed and have garnered significant attention over the past five years. Their extraordinary properties, including regulatable enzymatic activity, stability, and multifunctionality stemming from magnetic properties, photothermal conversion effects, cavitation effects, and relaxation efficiency, have made Co-based nanozymes a rising star. This review presents the first comprehensive profiling of the Co-based nanozymes in the chemistry, biology, and environmental sciences. The review begins by scrutinizing the various synthetic methods employed for Co-based nanozyme fabrication, such as template and sol-gel methods, highlighting their distinctive merits from a chemical standpoint. Furthermore, a detailed exploration of their wide-ranging applications in biosensing and biomedical therapeutics, as well as their contributions to environmental monitoring and remediation is provided. Notably, drawing inspiration from state-of-the-art techniques such as omics, a comprehensive analysis of Co-based nanozymes is undertaken, employing analogous statistical methodologies to provide valuable guidance. To conclude, a comprehensive outlook on the challenges and prospects for Co-based nanozymes is presented, spanning from microscopic physicochemical mechanisms to macroscopic clinical translational applications.
Collapse
Affiliation(s)
- Jingqi Li
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, P. R. China
- Aulin College, Northeast Forestry University, Harbin, 150040, P. R. China
| | - Xinda Cai
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, P. R. China
- Aulin College, Northeast Forestry University, Harbin, 150040, P. R. China
| | - Peng Jiang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, P. R. China
- Aulin College, Northeast Forestry University, Harbin, 150040, P. R. China
| | - Huayuan Wang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, P. R. China
- Aulin College, Northeast Forestry University, Harbin, 150040, P. R. China
| | - Shiwei Zhang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, P. R. China
- Aulin College, Northeast Forestry University, Harbin, 150040, P. R. China
| | - Tiedong Sun
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, P. R. China
- Aulin College, Northeast Forestry University, Harbin, 150040, P. R. China
| | - Chunxia Chen
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, P. R. China
- Aulin College, Northeast Forestry University, Harbin, 150040, P. R. China
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, P. R. China
| |
Collapse
|
4
|
House A, Cornick J, Butt Q, Guvendiren M. Elastomeric platform with surface wrinkling patterns to control cardiac cell alignment. J Biomed Mater Res A 2023; 111:1228-1242. [PMID: 36762538 DOI: 10.1002/jbm.a.37511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 12/07/2022] [Accepted: 01/29/2023] [Indexed: 02/11/2023]
Abstract
There is a growing interest in creating 2D cardiac tissue models that display native extracellular matrix (ECM) cues of the heart tissue. Cellular alignment alone is known to be a crucial cue for cardiac tissue development by regulating cell-cell and cell-ECM interactions. In this study, we report a simple and robust approach to create lamellar surface wrinkling patterns enabling spatial control of pattern dimensions with a wide range of pattern amplitude (A ≈ 2-55 μm) and wavelength (λ ≈ 35-100 μm). For human cardiomyocytes (hCMs) and human cardiac fibroblasts (hCFs), our results indicate that the degree of cellular alignment and pattern recognition are correlated with pattern A and λ. We also demonstrate fabrication of devices composed of micro-well arrays with user-defined lamellar patterns on the bottom surface of each well for high-throughput screening studies. Results from a screening study indicate that cellular alignment is strongly diminished with increasing seeding density. In another study, we show our ability to vary hCM/hCF seeding ratio for each well to create co-culture systems where seeding ratio is independent of cellular alignment.
Collapse
Affiliation(s)
- Andrew House
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| | - Jason Cornick
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| | - Quratulain Butt
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| | - Murat Guvendiren
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| |
Collapse
|
5
|
Zhou Y, Ping X, Guo Y, Heng BC, Wang Y, Meng Y, Jiang S, Wei Y, Lai B, Zhang X, Deng X. Assessing Biomaterial-Induced Stem Cell Lineage Fate by Machine Learning-Based Artificial Intelligence. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2210637. [PMID: 36756993 DOI: 10.1002/adma.202210637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/02/2023] [Indexed: 05/12/2023]
Abstract
Current functional assessment of biomaterial-induced stem cell lineage fate in vitro mainly relies on biomarker-dependent methods with limited accuracy and efficiency. Here a "Mesenchymal stem cell Differentiation Prediction (MeD-P)" framework for biomaterial-induced cell lineage fate prediction is reported. MeD-P contains a cell-type-specific gene expression profile as a reference by integrating public RNA-seq data related to tri-lineage differentiation (osteogenesis, chondrogenesis, and adipogenesis) of human mesenchymal stem cells (hMSCs) and a predictive model for classifying hMSCs differentiation lineages using the k-nearest neighbors (kNN) strategy. It is shown that MeD-P exhibits an overall accuracy of 90.63% on testing datasets, which is significantly higher than the model constructed based on canonical marker genes (80.21%). Moreover, evaluations of multiple biomaterials show that MeD-P provides accurate prediction of lineage fate on different types of biomaterials as early as the first week of hMSCs culture. In summary, it is demonstrated that MeD-P is an efficient and accurate strategy for stem cell lineage fate prediction and preliminary biomaterial functional evaluation.
Collapse
Affiliation(s)
- Yingying Zhou
- Department of Dental Materials and Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Xianfeng Ping
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Yusi Guo
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Boon Chin Heng
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Yijun Wang
- Department of Dental Materials and Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Yanze Meng
- Department of Dental Materials and Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Shengjie Jiang
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Yan Wei
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Binbin Lai
- Biomedical Engineering Department, Peking University, Beijing, 100191, P. R. China
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, 100034, P. R. China
| | - Xuehui Zhang
- Department of Dental Materials and Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Xuliang Deng
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Biomedical Engineering Department, Peking University, Beijing, 100191, P. R. China
| |
Collapse
|
6
|
Canullo L, Rakic M, Corvino E, Burton M, Krumbeck JA, Chittoor Prem A, Ravidà A, Ignjatović N, Sculean A, Menini M, Pesce P. Effect of argon plasma pre-treatment of healing abutments on peri-implant microbiome and soft tissue integration: a proof-of-concept randomized study. BMC Oral Health 2023; 23:27. [PMID: 36650477 PMCID: PMC9843976 DOI: 10.1186/s12903-023-02729-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 01/10/2023] [Indexed: 01/18/2023] Open
Abstract
PURPOSE Biofilm-free implant surface is ultimate prerequisite for successful soft and bone tissue integration. Objective of the study was to estimate the effects of argon plasma healing abutment pre-treatment (PT) on peri-implant soft-tissue phenotype (PiSP), inflammation, plaque accumulation and the microbiome (PiM) between non-treated (NPT) and treated (PT) abutments following 3-months healing period. The hypothesis was that cell-conductive and antimicrobial properties of PT would yield optimal conditions for soft tissue integration. MATERIAL AND METHODS Two months following second-phase surgery, microbiological and clinical parameters were assessed around thirty-six healing abutments with two types of microtopography, smooth surface (MACHINED) and ultrathin threaded microsurface (ROUGH). A two level randomization schema was used to achieve equal distribution and abutments were randomly divided into rough and machined groups, and then divided into PT and NPT groups. PiM was assessed using next-generation DNA sequencing. RESULTS PiM bacterial composition was highly diverse already two months post-implantation, consisting of key-stone pathogens, early and late colonizers, while the mycobiome was less diverse. PT was associated with lower plaque accumulation and inflammation without significant impact on PiSP, while in NPT clinical parameters were increased and associated with periopathogens. NPT mostly harbored late colonizers, while PT exerted higher abundance of early colonizers suggesting less advanced plaque formation. Interaction analysis in PT demonstrated S. mitis co-occurrence with pro-healthy Rothia dentocariosa and co-exclusion with Parvimonas micra, Porphyromonas endodontalis and Prevotella oris. PiSP parameters were generally similar between the groups, but significant association between PiM and keratinized mucosa width was observed in both groups, with remarkably more expressed diversity in NPT compared to PT. PT resulted in significantly lower BOP and PI around rough and machined abutments, respectively, without specific effect on PiM and PiSP. CONCLUSIONS PT contributed to significantly the less advanced biofilm accumulation and inflammation without specific effects on PiSP.
Collapse
Affiliation(s)
- Luigi Canullo
- grid.5734.50000 0001 0726 5157Department of Periodontology, University of Bern, Bern, Switzerland ,grid.5606.50000 0001 2151 3065Department of Surgical Sciences (DISC), University of Genoa, Genoa, Italy
| | - Mia Rakic
- grid.4795.f0000 0001 2157 7667ETEP (Etiology and Therapy of Periodontal Diseases) Research Group, University Complutense of Madrid, Madrid, Spain
| | - Emilio Corvino
- grid.8404.80000 0004 1757 2304University of Florence, Florence, Italy
| | - Maria Burton
- Zymo Research Corporation, 17062 Murphy Ave, Irvine, CA 92614 USA
| | - Janina A. Krumbeck
- Zymo Research Corporation, 17062 Murphy Ave, Irvine, CA 92614 USA ,Pangea Laboratory, 14762 Bentley Cir., Tustin, CA 92780 USA
| | | | | | - Nenad Ignjatović
- grid.419857.60000 0001 2221 9722Institute of Technical Science of the Serbian Academy of Sciences and Arts, 11000 Belgrade, Serbia
| | - Anton Sculean
- grid.5734.50000 0001 0726 5157Department of Periodontology, University of Bern, Bern, Switzerland
| | - Maria Menini
- grid.5606.50000 0001 2151 3065Department of Surgical Sciences (DISC), University of Genoa, Genoa, Italy
| | - Paolo Pesce
- grid.5606.50000 0001 2151 3065Department of Surgical Sciences (DISC), University of Genoa, Genoa, Italy
| |
Collapse
|
7
|
Zhong JX, Raghavan P, Desai TA. Harnessing Biomaterials for Immunomodulatory-Driven Tissue Engineering. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2022; 9:224-239. [PMID: 37333620 PMCID: PMC10272262 DOI: 10.1007/s40883-022-00279-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/08/2022] [Accepted: 09/07/2022] [Indexed: 11/29/2022]
Abstract
Abstract The immune system plays a crucial role during tissue repair and wound healing processes. Biomaterials have been leveraged to assist in this in situ tissue regeneration process to dampen the foreign body response by evading or suppressing the immune system. An emerging paradigm within regenerative medicine is to use biomaterials to influence the immune system and create a pro-reparative microenvironment to instigate endogenously driven tissue repair. In this review, we discuss recent studies that focus on immunomodulation of innate and adaptive immune cells for tissue engineering applications through four biomaterial-based mechanisms of action: biophysical cues, chemical modifications, drug delivery, and sequestration. These materials enable augmented regeneration in various contexts, including vascularization, bone repair, wound healing, and autoimmune regulation. While further understanding of immune-material interactions is needed to design the next generation of immunomodulatory biomaterials, these materials have already demonstrated great promise for regenerative medicine. Lay Summary The immune system plays an important role in tissue repair. Many biomaterial strategies have been used to promote tissue repair, and recent work in this area has looked into the possibility of doing repair by tuning. Thus, we examined the literature for recent works showcasing the efficacy of these approaches in animal models of injuries. In these studies, we found that biomaterials successfully tuned the immune response and improved the repair of various tissues. This highlights the promise of immune-modulating material strategies to improve tissue repair.
Collapse
Affiliation(s)
- Justin X. Zhong
- UC Berkeley – UCSF Graduate Program in Bioengineering, San Francisco, CA 94143 USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143 USA
| | - Preethi Raghavan
- UC Berkeley – UCSF Graduate Program in Bioengineering, San Francisco, CA 94143 USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143 USA
| | - Tejal A. Desai
- UC Berkeley – UCSF Graduate Program in Bioengineering, San Francisco, CA 94143 USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143 USA
- Department of Bioengineering, University of California, Berkeley, CA 94720 USA
- School of Engineering, Brown University, Providence, RI 02912 USA
| |
Collapse
|
8
|
Nanocellulose-based hydrogels as versatile drug delivery vehicles: A review. Int J Biol Macromol 2022; 222:830-843. [PMID: 36179866 DOI: 10.1016/j.ijbiomac.2022.09.214] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/19/2022] [Accepted: 09/24/2022] [Indexed: 11/22/2022]
Abstract
Hydrogels designed with nanocellulose (i.e. cellulose nanocrystals (CNC), cellulose nanofibrils (CNF), and bacterial cellulose (BC)) have significant advantages as drug carriers due to their environmentally-benign features and excellent properties. Nanocellulose hydrogels have been demonstrated to sustainably deliver various kinds of drugs via different routes of administration, in which nanocellulose significantly improves the hydrogel properties and tunes the drug releasing profile. This article comprehensively summarizes the recent research progress on nanocellulose hydrogels in drug delivery. We carefully assessed the gelation methods for nanocellulose hydrogel design and highlighted the influence of nanocellulose on hydrogel properties and drug release behaviors. In particular, it is the first time to summarize the research on nanocellulose hydrogel-based drug carriers regarding specific routes of administration. This work provides a critical review of nanocellulose-based hydrogels as drug delivery vehicles, and also underlines the outlook in this field, with the objective to inspire/prompt future work, especially the practical applications of nanocellulose hydrogels in designing controlled drug delivery systems.
Collapse
|
9
|
Kumar R. Materiomically Designed Polymeric Vehicles for Nucleic Acids: Quo Vadis? ACS APPLIED BIO MATERIALS 2022; 5:2507-2535. [PMID: 35642794 DOI: 10.1021/acsabm.2c00346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Despite rapid advances in molecular biology, particularly in site-specific genome editing technologies, such as CRISPR/Cas9 and base editing, financial and logistical challenges hinder a broad population from accessing and benefiting from gene therapy. To improve the affordability and scalability of gene therapy, we need to deploy chemically defined, economical, and scalable materials, such as synthetic polymers. For polymers to deliver nucleic acids efficaciously to targeted cells, they must optimally combine design attributes, such as architecture, length, composition, spatial distribution of monomers, basicity, hydrophilic-hydrophobic phase balance, or protonation degree. Designing polymeric vectors for specific nucleic acid payloads is a multivariate optimization problem wherein even minuscule deviations from the optimum are poorly tolerated. To explore the multivariate polymer design space rapidly, efficiently, and fruitfully, we must integrate parallelized polymer synthesis, high-throughput biological screening, and statistical modeling. Although materiomics approaches promise to streamline polymeric vector development, several methodological ambiguities must be resolved. For instance, establishing a flexible polymer ontology that accommodates recent synthetic advances, enforcing uniform polymer characterization and data reporting standards, and implementing multiplexed in vitro and in vivo screening studies require considerable planning, coordination, and effort. This contribution will acquaint readers with the challenges associated with materiomics approaches to polymeric gene delivery and offers guidelines for overcoming these challenges. Here, we summarize recent developments in combinatorial polymer synthesis, high-throughput screening of polymeric vectors, omics-based approaches to polymer design, barcoding schemes for pooled in vitro and in vivo screening, and identify materiomics-inspired research directions that will realize the long-unfulfilled clinical potential of polymeric carriers in gene therapy.
Collapse
Affiliation(s)
- Ramya Kumar
- Department of Chemical & Biological Engineering, Colorado School of Mines, 1613 Illinois St, Golden, Colorado 80401, United States
| |
Collapse
|
10
|
Cerqueira A, García-Arnáez I, Romero-Gavilán F, Azkargorta M, Elortza F, Martín de Llanos JJ, Carda C, Gurruchaga M, Goñi I, Suay J. Complex effects of Mg-biomaterials on the osteoblast cell machinery: A proteomic study. BIOMATERIALS ADVANCES 2022; 137:212826. [PMID: 35929259 DOI: 10.1016/j.bioadv.2022.212826] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/08/2022] [Accepted: 04/20/2022] [Indexed: 12/16/2022]
Abstract
The cell-biomaterial interface is highly complex; thousands of molecules and many processes participate in its formation. Growing demand for improved biomaterials has highlighted the need to understand the structure and functions of this interface. Proteomic methods offer a viable alternative to the traditional in vitro techniques for analyzing such systems. Magnesium is a promoter of cell adhesion and osteogenesis. Here, we used the LC-MS/MS to compare the protein expression profiles of human osteoblasts (HOb) exposed to sol-gel coatings without (MT) and with Mg (MT1.5Mg) for 1, 3, and 7 days. PANTHER, DAVID, and IPA databases were employed for protein identification and data analysis. Confocal microscopy and gene expression analysis were used for further characterization. Exposure to MT1.5Mg increased the HOb cell area and the expression of SP7, RUNX2, IBP3, COL3A1, MXRA8, and FBN1 genes. Proteomic analysis showed that MT1.5Mg affected the early osteoblast maturation (PI3/AKT, mTOR, ERK/MAPK), insulin metabolism, cell adhesion (integrin, FAK, actin cytoskeleton regulation) and oxidative stress pathways. Thus, the effects of Mg on cell adhesion and osteogenesis are rather complex, affecting several pathways rather than single processes. Our analysis also confirms the potential of proteomics in biomaterial characterization, showing a good correlation with in vitro results.
Collapse
Affiliation(s)
- Andreia Cerqueira
- Department of Industrial Systems Engineering and Design, Universitat Jaume I, Av. Vicent Sos Baynat s/n, 12071 Castellón de la Plana, Spain
| | - Iñaki García-Arnáez
- Department of Science and Technology of Polymers, University of the Basque Country, P. M. de Lardizábal, 3, 20018 San Sebastián, Spain
| | - Francisco Romero-Gavilán
- Department of Industrial Systems Engineering and Design, Universitat Jaume I, Av. Vicent Sos Baynat s/n, 12071 Castellón de la Plana, Spain.
| | - Mikel Azkargorta
- Proteomics Platform, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), CIBERehd, ProteoRed-ISCIII, Bizkaia Science and Technology Park, 48160 Derio, Spain
| | - Félix Elortza
- Proteomics Platform, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), CIBERehd, ProteoRed-ISCIII, Bizkaia Science and Technology Park, 48160 Derio, Spain
| | - José Javier Martín de Llanos
- Department of Pathology Medicine and Odontology, Medicine Faculty, University of Valencia, Av Blasco Ibáñez, 13, 46010, Valencia, Spain; Research Institute of the University Clinical Hospital of Valencia (INCLIVA), C. de Menéndez y Pelayo, 4, 46010, Valencia, Spain
| | - Carmen Carda
- Department of Pathology Medicine and Odontology, Medicine Faculty, University of Valencia, Av Blasco Ibáñez, 13, 46010, Valencia, Spain; Research Institute of the University Clinical Hospital of Valencia (INCLIVA), C. de Menéndez y Pelayo, 4, 46010, Valencia, Spain
| | - Mariló Gurruchaga
- Department of Science and Technology of Polymers, University of the Basque Country, P. M. de Lardizábal, 3, 20018 San Sebastián, Spain
| | - Isabel Goñi
- Department of Science and Technology of Polymers, University of the Basque Country, P. M. de Lardizábal, 3, 20018 San Sebastián, Spain
| | - Julio Suay
- Department of Industrial Systems Engineering and Design, Universitat Jaume I, Av. Vicent Sos Baynat s/n, 12071 Castellón de la Plana, Spain
| |
Collapse
|
11
|
Basu B, Gowtham N, Xiao Y, Kalidindi SR, Leong KW. Biomaterialomics: Data science-driven pathways to develop fourth-generation biomaterials. Acta Biomater 2022; 143:1-25. [PMID: 35202854 DOI: 10.1016/j.actbio.2022.02.027] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 12/12/2022]
Abstract
Conventional approaches to developing biomaterials and implants require intuitive tailoring of manufacturing protocols and biocompatibility assessment. This leads to longer development cycles, and high costs. To meet existing and unmet clinical needs, it is critical to accelerate the production of implantable biomaterials, implants and biomedical devices. Building on the Materials Genome Initiative, we define the concept 'biomaterialomics' as the integration of multi-omics data and high-dimensional analysis with artificial intelligence (AI) tools throughout the entire pipeline of biomaterials development. The Data Science-driven approach is envisioned to bring together on a single platform, the computational tools, databases, experimental methods, machine learning, and advanced manufacturing (e.g., 3D printing) to develop the fourth-generation biomaterials and implants, whose clinical performance will be predicted using 'digital twins'. While analysing the key elements of the concept of 'biomaterialomics', significant emphasis has been put forward to effectively utilize high-throughput biocompatibility data together with multiscale physics-based models, E-platform/online databases of clinical studies, data science approaches, including metadata management, AI/ Machine Learning (ML) algorithms and uncertainty predictions. Such integrated formulation will allow one to adopt cross-disciplinary approaches to establish processing-structure-property (PSP) linkages. A few published studies from the lead author's research group serve as representative examples to illustrate the formulation and relevance of the 'Biomaterialomics' approaches for three emerging research themes, i.e. patient-specific implants, additive manufacturing, and bioelectronic medicine. The increased adaptability of AI/ML tools in biomaterials science along with the training of the next generation researchers in data science are strongly recommended. STATEMENT OF SIGNIFICANCE: This leading opinion review paper emphasizes the need to integrate the concepts and algorithms of the data science with biomaterials science. Also, this paper emphasizes the need to establish a mathematically rigorous cross-disciplinary framework that will allow a systematic quantitative exploration and curation of critical biomaterials knowledge needed to drive objectively the innovation efforts within a suitable uncertainty quantification framework, as embodied in 'biomaterialomics' concept, which integrates multi-omics data and high-dimensional analysis with artificial intelligence (AI) tools, like machine learning. The formulation of this approach has been demonstrated for patient-specific implants, additive manufacturing, and bioelectronic medicine.
Collapse
|
12
|
Tuvshindorj U, Trouillet V, Vasilevich A, Koch B, Vermeulen S, Carlier A, Alexander MR, Giselbrecht S, Truckenmüller R, de Boer J. The Galapagos Chip Platform for High-Throughput Screening of Cell Adhesive Chemical Micropatterns. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2105704. [PMID: 34985808 DOI: 10.1002/smll.202105704] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/04/2021] [Indexed: 06/14/2023]
Abstract
In vivo cells reside in a complex extracellular matrix (ECM) that presents spatially distributed biochemical and -physical cues at the nano- to micrometer scales. Chemical micropatterning is successfully used to generate adhesive islands to control where and how cells attach and restore cues of the ECM in vitro. Although chemical micropatterning has become a powerful tool to study cell-material interactions, only a fraction of the possible micropattern designs was covered so far, leaving many other possible designs still unexplored. Here, a high-throughput screening platform called "Galapagos chip" is developed. It contains a library of 2176 distinct subcellular chemical patterns created using mathematical algorithms and a straightforward UV-induced two-step surface modification. This approach enables the immobilization of ligands in geometrically defined regions onto cell culture substrates. To validate the system, binary RGD/polyethylene glycol patterns are prepared on which human mesenchymal stem cells are cultured, and the authors observe how different patterns affect cell and organelle morphology. As proof of concept, the cells are stained for the mechanosensitive YAP protein, and, using a machine-learning algorithm, it is demonstrated that cell shape and YAP nuclear translocation correlate. It is concluded that the Galapagos chip is a versatile platform to screen geometrical aspects of cell-ECM interaction.
Collapse
Affiliation(s)
- Urandelger Tuvshindorj
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, 6229 ER, The Netherlands
- Department of Biomedical Engineering and Institute, for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, 5600 MB, The Netherlands
| | - Vanessa Trouillet
- Institute for Applied Materials and Karlsruhe Nano Micro Facility, Karlsruhe Institute of Technology, 76344, Eggenstein-Leopoldshafen, Germany
| | - Aliaksei Vasilevich
- Department of Biomedical Engineering and Institute, for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, 5600 MB, The Netherlands
| | - Britta Koch
- Advanced Materials and Healthcare Technologies Division, The School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Steven Vermeulen
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, 6229 ER, The Netherlands
| | - Aurélie Carlier
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, 6229 ER, The Netherlands
| | - Morgan R Alexander
- Advanced Materials and Healthcare Technologies Division, The School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Stefan Giselbrecht
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, 6229 ER, The Netherlands
| | - Roman Truckenmüller
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, 6229 ER, The Netherlands
| | - Jan de Boer
- Department of Biomedical Engineering and Institute, for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, 5600 MB, The Netherlands
| |
Collapse
|
13
|
Sari B, Isik M, Eylem CC, Kilic C, Okesola BO, Karakaya E, Emregul E, Nemutlu E, Derkus B. Omics Technologies for High-Throughput-Screening of Cell-Biomaterial Interactions. Mol Omics 2022; 18:591-615. [DOI: 10.1039/d2mo00060a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Recent research effort in biomaterial development has largely focused on engineering bio-instructive materials to stimulate specific cell signaling. Assessing the biological performance of these materials using time-consuming and trial-and-error traditional...
Collapse
|
14
|
Ansar R, Saqib S, Mukhtar A, Niazi MBK, Shahid M, Jahan Z, Kakar SJ, Uzair B, Mubashir M, Ullah S, Khoo KS, Lim HR, Show PL. Challenges and recent trends with the development of hydrogel fiber for biomedical applications. CHEMOSPHERE 2022; 287:131956. [PMID: 34523459 DOI: 10.1016/j.chemosphere.2021.131956] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/12/2021] [Accepted: 08/18/2021] [Indexed: 06/13/2023]
Abstract
Hydrogel is the most emblematic soft material which possesses significantly tunable and programmable characteristics. Polymer hydrogels possess significant advantages including, biocompatible, simple, reliable and low cost. Therefore, research on the development of hydrogel for biomedical applications has been grown intensely. However, hydrogel development is challenging and required significant effort before the application at an industrial scale. Therefore, the current work focused on evaluating recent trends and issues with hydrogel development for biomedical applications. In addition, the hydrogel's development methodology, physicochemical properties, and biomedical applications are evaluated and benchmarked against the reported literature. Later, biomedical applications of the nano-cellulose-based hydrogel are considered and critically discussed. Based on a detailed review, it has been found that the surface energy, intermolecular interactions, and interactions of hydrogel adhesion forces are major challenges that contribute to the development of hydrogel. In addition, compared to other hydrogels, nanocellulose hydrogels demonstrated higher potential for drug delivery, 3D cell culture, diagnostics, tissue engineering, tissue therapies and gene therapies. Overall, nanocellulose hydrogel has the potential for commercialization for different biomedical applications.
Collapse
Affiliation(s)
- Reema Ansar
- Department of Chemical Engineering, University of Gujrat, 50700, Pakistan.
| | - Sidra Saqib
- Department of Chemical Engineering, COMSATS University Islamabad, Lahore Campus, 54000, Lahore, Pakistan.
| | - Ahmad Mukhtar
- Department of Chemical Engineering, NFC Institute of Engineering and Fertilizer Research, Jaranwala Road, 38000, Faisalabad, Pakistan.
| | - Muhammad Bilal Khan Niazi
- School of Chemical and Materials Engineering, National University of Sciences and Technology, Islamabad, Pakistan.
| | - Muhammad Shahid
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, 38000, Pakistan.
| | - Zaib Jahan
- School of Chemical and Materials Engineering, National University of Sciences and Technology, Islamabad, Pakistan.
| | - Salik Javed Kakar
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Sector H-12, Islamabad, Pakistan.
| | - Bushra Uzair
- Department of Biological Sciences, International Islamic University Islamabad, Islamabad, Pakistan.
| | - Muhammad Mubashir
- Department of Petroleum Engineering, School of Engineering, Asia Pacific University of Technology and Innovation, 57000, Kuala Lumpur, Malaysia.
| | - Sami Ullah
- Department of Chemistry, College of Science, King Khalid University, Abha, Saudi Arabia.
| | - Kuan Shiong Khoo
- Department of Chemical and Environmental Engineering, Faculty Science and Engineering, University of Nottingham, Malaysia, 43500, Semenyih, Selangor Darul Ehsan, Malaysia.
| | - Hooi Ren Lim
- Department of Chemical and Environmental Engineering, Faculty Science and Engineering, University of Nottingham, Malaysia, 43500, Semenyih, Selangor Darul Ehsan, Malaysia.
| | - Pau Loke Show
- Department of Chemical and Environmental Engineering, Faculty Science and Engineering, University of Nottingham, Malaysia, 43500, Semenyih, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
15
|
Palmieri V, De Spirito M, Papi M. Nanofeatures of orthopedic implant surfaces. Nanomedicine (Lond) 2021; 16:1733-1736. [PMID: 34196227 DOI: 10.2217/nnm-2021-0118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Valentina Palmieri
- Istituto dei Sistemi Complessi, CNR, Via dei Taurini 19, Rome, 00185, Italy.,Fondazione Policlinico Universitario 'A Gemelli' IRCSS, Rome, Italy
| | - Marco De Spirito
- Fondazione Policlinico Universitario 'A Gemelli' IRCSS, Rome, Italy.,Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Massimiliano Papi
- Fondazione Policlinico Universitario 'A Gemelli' IRCSS, Rome, Italy.,Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
16
|
Yang J, Qin H, Chai Y, zhang P, Chen Y, Yang K, Qin M, Zhang Y, Xia H, Ren L, Yu B. Molecular mechanisms of osteogenesis and antibacterial activity of Cu-bearing Ti alloy in a bone defect model with infection in vivo. J Orthop Translat 2021; 27:77-89. [PMID: 33437640 PMCID: PMC7779545 DOI: 10.1016/j.jot.2020.10.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 10/03/2020] [Accepted: 10/08/2020] [Indexed: 01/28/2023] Open
Abstract
OBJECTIVE The antibacterial activity of copper (Cu)-alloy biomaterials has shown a great potential in clinical application. Here, we evaluated the osteogenesis and antibacterial effects of Ti6Al4V-6.5wt%Cu alloy in an in vivo model of infected bone defects and determine their responsible proteins and pathways using proteomics. METHODS After bone defects were filled with Ti6Al4V and Ti6Al4V-6.5wt%Cu implants for 6 week, the tissue and bone samples around the implants were harvested for radiographic, micro-CT, histological, and bone-related gene expression analyses. An iTRAQ-based protein identification/quantification approach was used to analyze the osteogenic and antibacterial effects of Ti6Al4V-6.5wt%Cu alloy. RESULTS Imaging and histological results showed Ti6Al4V alloy induced a stronger inflammatory response than Ti6Al4V-6.5wt%Cu alloy; imaging results and osteogenic protein levels showed Ti6Al4V-6.5wt%Cu alloy exerted a stronger osteogenic effect. In vitro experiment, we found the Ti6Al4V-6.5wt%Cu had significant antibacterial effects and inhibited the activity of Staphylococcus aureus in the early stage. In addition, the bacterial biofilm formed in Ti6Al4V-6.5wt%Cu group was significantly lower than that in Ti6Al4V group. Proteomic screening of 4279 proteins resulted in 35 differentially expressed proteins for further examination which were mainly associated with the cellular process, metabolic process, stimulus response, and cellular component organization. In further exploration of the mechanism of osteogenic mineralization of Ti6Al4V-6.5wt%Cu alloy, we found out SDC4 and AGRN were the top two target proteins associated with osteogenic differentiation and bone mineralization. CONCLUSION Ti6Al4V-6.5wt%Cu alloy shows a great potential as a bone implant material due to its positive effects against bacterial infection and on bone formation. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE At present, titanium alloys and other non-antibacterial metal materials are used in orthopedic internal fixation operations. Our study demonstrates that Ti6Al4V-6.5wt%Cu alloy has good antibacterial and osteogenic effects in vivo and in vitro. This means that Ti6Al4V-6.5wt%Cu alloy may become a new kind of antimicrobial metallic material as internal fixation material to continuously exert its antimicrobial effects and reduce the infection rate after clinical internal fixation.
Collapse
Key Words
- AGRN, Agrin
- ALP, alkaline phosphatase
- Antibacterial
- BV, bone volume
- Bone defect
- DEPs, differentially expressed proteins
- EDTA, Ethylene Diamine Tetraacetic Acid
- ESI, Electrospray Ionization
- LC, Liquid Chromatography
- OCN, osteocalcin
- OPN, osteopontin
- Osteogenesis
- PPI, protein-to-protein interacting
- S. Aureus, staphylococcus aureus
- SCX, Strong Cation Exchange
- SDC4, Syndecan 4
- SEM, scanning electron microscope
- TV, tissue volume
- Tb.N, trabecular number
- Tb.Sp, trabecular separation
- Tb.Th, trabecular thickness
- Ti6Al4V-6.5wt%Cu alloy
- UV, ultraviolet
- XRD, X-Ray Diffraction
- cfu, colony-forming unit
- hBMSCs, human bone marrow stromal cells
- iTRAQ, isobaric Tags for Relative and Absolute Quantitation
- isobaric tags for relative and absolute quantification(iTRAQ) analysis
- micro-CT, microcomputed tomography
- pAGC, predictive Automatic Gain Control
Collapse
Affiliation(s)
- Jun Yang
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
- Department of Orthopaedics, Guangzhou General Hospital of Guangzhou Military Command of PLA, Guangdong Key Lab of Orthopaedic Technology and Implant Materials, Key Laboratory of Trauma & Tissue Repair of Tropical Area of PLA, Guangzhou 510010, China
| | - Hanjun Qin
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yu Chai
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Ping zhang
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yirong Chen
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Ke Yang
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, China
| | - Min Qin
- School of Public Health, Experimental Teaching Center of Preventive Medicine, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yifang Zhang
- Editorial Office, Chinese Journal of Orthopaedic Trauma, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Hong Xia
- Department of Orthopaedics, Guangzhou General Hospital of Guangzhou Military Command of PLA, Guangdong Key Lab of Orthopaedic Technology and Implant Materials, Key Laboratory of Trauma & Tissue Repair of Tropical Area of PLA, Guangzhou 510010, China
| | - Ling Ren
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, China
| | - Bin Yu
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| |
Collapse
|
17
|
Liu P, Zhang S, Gao L, Wang H, Guo J, Huang J, Liu L. Progress in Application of Carrageenan Hydrogel in Biomedicine. J PHOTOPOLYM SCI TEC 2021. [DOI: 10.2494/photopolymer.34.615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Panpan Liu
- School of Chemical Engineering and Technology, North University of China
| | - Shuli Zhang
- School of Chemical Engineering and Technology, North University of China
| | - Li Gao
- School of Chemical Engineering and Technology, North University of China
| | - Haibin Wang
- School of Chemical Engineering and Technology, North University of China
| | - Jianfeng Guo
- School of Chemical Engineering and Technology, North University of China
| | - Jingjing Huang
- School of Chemical Engineering and Technology, North University of China
| | - Linlin Liu
- School of Chemical Engineering and Technology, North University of China
| |
Collapse
|
18
|
Lei R, Kumar S. Getting the big picture of cell-matrix interactions: High-throughput biomaterial platforms and systems-level measurements. CURRENT OPINION IN SOLID STATE & MATERIALS SCIENCE 2020; 24:100871. [PMID: 33244294 PMCID: PMC7685248 DOI: 10.1016/j.cossms.2020.100871] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Living cells interact with the extracellular matrix (ECM) in a complex and reciprocal manner. Much has been learned over the past few decades about cell-ECM interactions from targeted studies in which a specific matrix parameter (e.g. stiffness, adhesivity) has been varied across a few discrete values, or in which the level or activity of a protein is controlled in an isolated fashion. As the field moves forward, there is growing interest in addressing cell-matrix interactions from a systems perspective, which has spurred a new generation of matrix platforms capable of interrogating multiple ECM inputs in a combinatorial and parallelized fashion. Efforts are also actively underway to integrate specialized, synthetic ECM platforms with global measures of cell behaviors, including at the transcriptomic, proteomic and epigenomic levels. Here we review recent advances in both areas. We describe how new combinatorial ECM technologies are revealing unexpected crosstalk and nonlinearity in the relationship between cell phenotype and matrix properties. Similarly, efforts to integrate "omics" measurements with synthetic ECM platforms are illuminating how ECM properties can control cell biology in surprising and functionally important ways. We expect that advances in both areas will deepen the field's understanding of cell-ECM interactions and offer valuable insight into the design of biomaterials for specific biomedical applications.
Collapse
Affiliation(s)
- Ruoxing Lei
- Department of Chemistry, University of California, Berkeley, CA, 94720
- Department of Bioengineering, University of California, Berkeley, CA, 94720
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, CA, 94720
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, 94720
| |
Collapse
|
19
|
Subhedar A, Bhadauria S, Ahankari S, Kargarzadeh H. Nanocellulose in biomedical and biosensing applications: A review. Int J Biol Macromol 2020; 166:587-600. [PMID: 33130267 DOI: 10.1016/j.ijbiomac.2020.10.217] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/20/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022]
Abstract
Cellulose is abundant in the nature and nanocellulose (NC) in particular is regarded as a credible green substrate to be used in bio nanocomposites for various applications. NC exhibits excellent mechanical reinforcement properties comparable to conventionally used materials due to its high specific surface area and tunable surface chemistry. Additionally, low toxicity, biodegradability and biocompatibility of NC deem it a promising material for use in different biomedical applications. In this review, we highlight the biomedical applications of NC based hydrogels and aerogels/nanocomposites and advancements of their employment in the areas of wound dressing, drug delivery, tissue engineering, scaffolds and biomedical implants. This review also explores the recent use of NC in making biosensors for the detection of cholesterol, various enzymes and diseases, heavy metal ions in human sweat and urine, and for general health monitoring.
Collapse
Affiliation(s)
- Aditya Subhedar
- School of Mechanical Engineering, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Swarnim Bhadauria
- School of Mechanical Engineering, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Sandeep Ahankari
- School of Mechanical Engineering, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India.
| | - Hanieh Kargarzadeh
- Center of Molecular and Macromolecular Studies, Polish Academy of Sciences, Seinkiewicza 112, 90-363 Lodz, Poland
| |
Collapse
|
20
|
Comprehensive Biological Evaluation of Biomaterials Used in Spinal and Orthopedic Surgery. MATERIALS 2020; 13:ma13214769. [PMID: 33114571 PMCID: PMC7672648 DOI: 10.3390/ma13214769] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/16/2020] [Accepted: 10/20/2020] [Indexed: 12/03/2022]
Abstract
Biological acceptance is one of the most important aspects of a biomaterial and forms the basis for its clinical use. The aim of this study was a comprehensive biological evaluation (cytotoxicity test, bacterial colonization test, blood platelets adhesion test and transcriptome and proteome analysis of Saos-2 cells after contact with surface of the biomaterial) of biomaterials used in spinal and orthopedic surgery, namely, Ti6Al4V ELI (Extra Low Interstitials), its modified version obtained as a result of melting by electron beam technology (Ti6Al4V ELI-EBT), polyether ether ketone (PEEK) and polished medical steel American Iron and Steel Institute (AISI) 316L (the reference material). Biological tests were carried out using the osteoblasts-like cells (Saos-2, ATCC HTB-85) and bacteria Escherichia coli (DH5α). Results showed lack of cytotoxicity of all materials and the surfaces of both Ti6Al4V ELI and PEEK exhibit a significantly higher resistance to colonization with E. coli cells, while the more porous surface of the same titanium alloy produced by electron beam technology (EBT) is more susceptible to microbial colonization than the control surface of polished medical steel. None of the tested materials showed high toxicity in relation to E. coli cells. Susceptibility to platelet adhesion was very high for polished medical steel AISI 316L, whilst much lower for the other biomaterials and can be ranked from the lowest to the highest as follows: PEEK < Ti6Al4V ELI < Ti6Al4V ELI-EBT. The number of expressed genes in Saos-2 cells exposed to contact with the examined biomaterials reached 9463 genes in total (ranging from 8455 genes expressed in cells exposed to ELI to 9160 genes in cells exposed to PEEK). Whereas the number of differentially expressed proteins detected on two-dimensional electrophoresis gels in Saos-2 cells after contact with the examined biomaterials was 141 for PEEK, 223 for Ti6Al4V ELI and 133 for Ti6Al4V ELI-EBT. Finally, 14 proteins with altered expression were identified by mass spectrometry. In conclusion, none of the tested biomaterials showed unsatisfactory levels of cytotoxicity. The gene and protein expression analysis, that represents a completely new approach towards characterization of these biomaterials, showed that the polymer PEEK causes much more intense changes in gene and protein expression and thus influences cell metabolism.
Collapse
|
21
|
Hall MS, Decker JT, Shea LD. Towards systems tissue engineering: Elucidating the dynamics, spatial coordination, and individual cells driving emergent behaviors. Biomaterials 2020; 255:120189. [PMID: 32569865 PMCID: PMC7396312 DOI: 10.1016/j.biomaterials.2020.120189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 04/20/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022]
Abstract
Biomaterial systems have enabled the in vitro production of complex, emergent tissue behaviors that were not possible with conventional two-dimensional culture systems, allowing for analysis of both normal development and disease processes. We propose that the path towards developing the design parameters for biomaterial systems lies with identifying the molecular drivers of emergent behavior through leveraging technological advances in systems biology, including single cell omics, genetic engineering, and high content imaging. This growing research opportunity at the intersection of the fields of tissue engineering and systems biology - systems tissue engineering - can uniquely interrogate the mechanisms by which complex tissue behaviors emerge with the potential to capture the contribution of i) dynamic regulation of tissue development and dysregulation, ii) single cell heterogeneity and the function of rare cell types, and iii) the spatial distribution and structure of individual cells and cell types within a tissue. By leveraging advances in both biological and materials data science, systems tissue engineering can facilitate the identification of biomaterial design parameters that will accelerate basic science discovery and translation.
Collapse
Affiliation(s)
- Matthew S Hall
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Joseph T Decker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
22
|
Reiss RA, Lowe TC, Sena JA, Makhnin O, Connick MC, Illescas PE, Davis CF. Bio-activating ultrafine grain titanium: RNA sequencing reveals enhanced mechano-activation of osteoconduction on nanostructured substrates. PLoS One 2020; 15:e0237463. [PMID: 32970688 PMCID: PMC7514099 DOI: 10.1371/journal.pone.0237463] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/27/2020] [Indexed: 02/02/2023] Open
Abstract
Titanium is essentially absent from biological systems yet reliably integrates into bone. To achieve osseointegration, titanium must activate biological processes without entering cells, defining it as a bio-activating material. Nanostructuring bulk titanium reduces grain size, increases strength, and improves other quantifiable physical properties, including cytocompatibility. The biological processes activated by increasing grain boundary availability were detected with total RNA-sequencing in mouse pre-osteoblasts grown for 72 hours on nanometrically smooth substrates of either coarse grain or nanostructured ultrafine grain titanium. The average grain boundary length under cells on the conventional coarse grain substrates is 273.0 μm, compared to 70,881.5 μm for cells adhered to the nanostructured ultrafine grain substrates; a 260-fold difference. Cells on both substrates exhibit similar expression profiles for genes whose products are critical for mechanosensation and transduction of cues that trigger osteoconduction. Biological process Gene Ontology term enrichment analysis of differentially expressed genes reveals that cell cycle, chromatin modification, telomere maintenance, and RNA metabolism processes are upregulated on ultrafine grain titanium. Processes related to immune response, including apoptosis, are downregulated. Tumor-suppressor genes are upregulated while tumor-promoting genes are downregulated. Upregulation of genes involved in chromatin remodeling and downregulation of genes under the control of the peripheral circadian clock implicate both processes in the transduction of mechanosensory information. Non-coding RNAs may also play a role in the response. Merging transcriptomics with well-established mechanobiology principles generates a unified model to explain the bio-activating properties of titanium. The modulation of processes is accomplished through chromatin remodeling in which the nucleus responds like a rheostat to grain boundary concentration. This convergence of biological and materials science reveals a pathway toward understanding the biotic-abiotic interface and will inform the development of effective bio-activating and bio-inactivating materials.
Collapse
Affiliation(s)
- Rebecca A. Reiss
- Biology Department, New Mexico Institution of Mining and Technology, Socorro, New Mexico, United States of America
| | - Terry C. Lowe
- George S. Ansell Department of Metallurgical and Materials Engineering, Colorado School of Mines, Golden, Colorado, United States of America
| | - Johnny A. Sena
- National Center for Genome Resources, Santa Fe, New Mexico, United States of America
| | - Oleg Makhnin
- Mathematics Department, New Mexico Institute of Mining and Technology, Socorro, New Mexico, United States of America
| | - Melanie C. Connick
- Biology Department, New Mexico Institution of Mining and Technology, Socorro, New Mexico, United States of America
| | - Patrick E. Illescas
- Biology Department, New Mexico Institution of Mining and Technology, Socorro, New Mexico, United States of America
| | - Casey F. Davis
- George S. Ansell Department of Metallurgical and Materials Engineering, Colorado School of Mines, Golden, Colorado, United States of America
| |
Collapse
|
23
|
Gholami P, Khataee A, Bhatnagar A. Environmentally superior cleaning of diatom frustules using sono-Fenton process: Facile fabrication of nanoporous silica with homogeneous morphology and controlled size. ULTRASONICS SONOCHEMISTRY 2020; 64:105044. [PMID: 32146334 DOI: 10.1016/j.ultsonch.2020.105044] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 01/28/2020] [Accepted: 02/24/2020] [Indexed: 06/10/2023]
Abstract
Existing techniques for the preparation of silica structures from diatom cells include cleaning of frustules through baking at high temperature and oxidant cleaning using concentrated sulfuric acid, hydrogen peroxide, nitric acid, or sodium dodecyl sulfate (SDS)/ethylenediaminetetraacetic acid (EDTA). In this study, sono-Fenton (SF) process was examined to prepare nanoporous silica through cleaning diatom frustules, while preserving their structural features. Single colonies of Cyclotella sp. were cultivated in batch mode f/2-enriched seawater. Combination of Fenton process with ultrasonication was found to be more efficient than the sum of individual processes in the removal of organic compounds from Cyclotella sp. structure. The optimized amounts of operational parameters were determined as suspension pH of 3, diatom cell density of 4.8 × 105 cell mL-1, H2O2 concentration of 60 mM, Fe2+ concentration of 15 mM, ultrasound irradiation power of 400 W and the temperature of 45 °C. The results of energy-dispersive X-ray spectroscopy (EDX) and thermal gravimetry (TG) analyses proved that organic materials covering the cell wall were significantly removed from the frustules through SF process. Scanning electron microscopy (SEM) images showed that after SF treatment, silica nanostructures were produced having uniform pores less than 15 nm in diameter. N2 adsorption-desorption isotherms demonstrated that almost non-porous structure of diatom frustules became mesoporous during removing the organic matrix. Lipids, amino acids, carbohydrates and organic acids or their oxidized products were identified using GC-MS analysis as the main organic compounds released from diatom cells to the solution after SF treatment. Treated frustules exhibited adsorption capability of 91.2 mg/g for Methylene Blue, which was almost 2.5 times higher than that of untreated frustules (34.8 mg/g).
Collapse
Affiliation(s)
- Peyman Gholami
- Research Laboratory of Advanced Water and Wastewater Treatment Processes, Department of Applied Chemistry, Faculty of Chemistry, University of Tabriz, 51666-16471 Tabriz, Iran; Department of Environmental and Biological Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Alireza Khataee
- Research Laboratory of Advanced Water and Wastewater Treatment Processes, Department of Applied Chemistry, Faculty of Chemistry, University of Tabriz, 51666-16471 Tabriz, Iran; Department of Materials Science and Nanotechnology Engineering, Faculty of Engineering, Near East University, 99138, Nicosia, TRNC, Mersin 10, Turkey; Institute of Research and Development, Duy Tan University, Da Nang 550000, Viet Nam.
| | - Amit Bhatnagar
- Department of Environmental and Biological Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| |
Collapse
|
24
|
Mullis AS, Jacobson SJ, Narasimhan B. High-Throughput Synthesis and Screening of Rapidly Degrading Polyanhydride Nanoparticles. ACS COMBINATORIAL SCIENCE 2020; 22:172-183. [PMID: 32125826 DOI: 10.1021/acscombsci.9b00162] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Combinatorial techniques can accelerate the discovery and development of polymeric nanodelivery devices by pairing high-throughput synthesis with rapid materials characterization. Biodegradable polyanhydrides demonstrate tunable release, high cellular internalization, and dose sparing properties when used as nanodelivery devices. This nanoparticle platform shows promising potential for small molecule drug delivery, but the pace of understanding and rational design of these nanomedicines is limited by the low throughput of conventional characterization. This study reports the use of a high-throughput method to synthesize libraries of a newly synthesized, rapidly eroding polyanhydride copolymer based on 1,8-bis(p-carboxyphenoxy)-3,6-dioxaoctane (CPTEG) and sebacic acid (SA) monomers. The high-throughput method enabled efficient screening of copolymer microstructure, revealing weak block-type and alternating architectures. The high-throughput method was adapted to synthesize nanoparticle libraries encapsulating hydrophobic model drugs. Drug release from these nanoparticles was rapid, with a majority of the payload released within 3 days. Drug release was dramatically slowed at acidic pH, which could be useful for oral drug delivery. Rhodamine B (RhoB) release kinetics generally followed patterns of polymer erosion kinetics, while Coomassie brilliant blue (CBB) released the fastest from the slowest degrading polymer chemistry and vice versa. These differences in trends between copolymer chemistry and release kinetics were hypothesized to arise from differences in mixing thermodynamics. A high-throughput method was developed to synthesize polymer-drug film libraries and characterize mixing thermodynamics by melting point depression. Rhodamine B had a negative χ for all copolymers with <30 mol % CPTEG tested, indicating a tendency toward miscibility. By contrast, CBB χ increased, eventually becoming positive near 15:85 CPTEG:SA, with increasing CPTEG content. This indicates an increasing tendency toward phase separation in CPTEG-rich copolymers. These in vitro results screening polymer-drug interactions showed good agreement with in silico predictions from Hansen solubility parameter estimation and were able to explain the observed differences in model drug release trends.
Collapse
Affiliation(s)
- Adam S. Mullis
- Department of Chemical and Biological Engineering and Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Sarah J. Jacobson
- Department of Chemical and Biological Engineering and Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering and Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| |
Collapse
|
25
|
Spencer P, Ye Q, Song L, Parthasarathy R, Boone K, Misra A, Tamerler C. Threats to adhesive/dentin interfacial integrity and next generation bio-enabled multifunctional adhesives. J Biomed Mater Res B Appl Biomater 2019; 107:2673-2683. [PMID: 30895695 PMCID: PMC6754319 DOI: 10.1002/jbm.b.34358] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 02/07/2019] [Accepted: 02/20/2019] [Indexed: 12/27/2022]
Abstract
Nearly 100 million of the 170 million composite and amalgam restorations placed annually in the United States are replacements for failed restorations. The primary reason both composite and amalgam restorations fail is recurrent decay, for which composite restorations experience a 2.0-3.5-fold increase compared to amalgam. Recurrent decay is a pernicious problem-the standard treatment is replacement of defective composites with larger restorations that will also fail, initiating a cycle of ever-larger restorations that can lead to root canals, and eventually, to tooth loss. Unlike amalgam, composite lacks the inherent capability to seal discrepancies at the restorative material/tooth interface. The low-viscosity adhesive that bonds the composite to the tooth is intended to seal the interface, but the adhesive degrades, which can breach the composite/tooth margin. Bacteria and bacterial by-products such as acids and enzymes infiltrate the marginal gaps and the composite's inability to increase the interfacial pH facilitates cariogenic and aciduric bacterial outgrowth. Together, these characteristics encourage recurrent decay, pulpal damage, and composite failure. This review article examines key biological and physicochemical interactions involved in the failure of composite restorations and discusses innovative strategies to mitigate the negative effects of pathogens at the adhesive/dentin interface. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B:2466-2475, 2019.
Collapse
Affiliation(s)
- Paulette Spencer
- Institute for Bioengineering Research, School of Engineering, University of Kansas, 1530 W. 15th Street, Lawrence, KS 66045-7609, USA
- Department of Mechanical Engineering, University of Kansas,1530 W. 15th Street, Lawrence, KS 66045-7609, USA
| | - Qiang Ye
- Institute for Bioengineering Research, School of Engineering, University of Kansas, 1530 W. 15th Street, Lawrence, KS 66045-7609, USA
| | - Linyong Song
- Institute for Bioengineering Research, School of Engineering, University of Kansas, 1530 W. 15th Street, Lawrence, KS 66045-7609, USA
| | - Ranganathan Parthasarathy
- Department of Civil Engineering, Tennessee State University, 3500 John A Merritt Blvd, Nashville, TN 37209, USA
| | - Kyle Boone
- Institute for Bioengineering Research, School of Engineering, University of Kansas, 1530 W. 15th Street, Lawrence, KS 66045-7609, USA
| | - Anil Misra
- Institute for Bioengineering Research, School of Engineering, University of Kansas, 1530 W. 15th Street, Lawrence, KS 66045-7609, USA
- Department of Civil Engineering, University of Kansas, 1530 W. 15th Street, Lawrence, KS 66045-7609, USA
| | - Candan Tamerler
- Institute for Bioengineering Research, School of Engineering, University of Kansas, 1530 W. 15th Street, Lawrence, KS 66045-7609, USA
- Department of Mechanical Engineering, University of Kansas,1530 W. 15th Street, Lawrence, KS 66045-7609, USA
| |
Collapse
|
26
|
Ouni E, Vertommen D, Amorim CA. The Human Ovary and Future of Fertility Assessment in the Post-Genome Era. Int J Mol Sci 2019; 20:E4209. [PMID: 31466236 PMCID: PMC6747278 DOI: 10.3390/ijms20174209] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 08/25/2019] [Accepted: 08/27/2019] [Indexed: 12/27/2022] Open
Abstract
Proteomics has opened up new avenues in the field of gynecology in the post-genome era, making it possible to meet patient needs more effectively and improve their care. This mini-review aims to reveal the scope of proteomic applications through an overview of the technique and its applications in assisted procreation. Some of the latest technologies in this field are described in order to better understand the perspectives of its clinical applications. Proteomics seems destined for a promising future in gynecology, more particularly in relation to the ovary. Nevertheless, we know that reproductive biology proteomics is still in its infancy and major technical and ethical challenges must first be overcome.
Collapse
Affiliation(s)
- Emna Ouni
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Didier Vertommen
- PHOS Unit, Institut de Duve, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Christiani A Amorim
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium.
| |
Collapse
|
27
|
Das D, Noh I. Overviews of Biomimetic Medical Materials. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1064:3-24. [PMID: 30471023 DOI: 10.1007/978-981-13-0445-3_1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
This chapter describes the overviews of biomimetic medical materials which covers innovation and significance of terminology, diverse fabrication methods, and technologies ranges from nanotechnology to 3D printing to develop biomimetic materials for medical applications. It also depicts specific fundamental characteristics required for a material to be a model biomimetic material for particular medical application. It basically outlines current statuses of biomimetic medical materials used for tissue engineering and regenerative medicine, drug/protein delivery, bioimaging, biosensing, and 3D bioprinting technology. It also illustrates the effect of functionalization of a material through chemical and biological approaches towards different applications. Not only, the key properties and potential applications of the biomimetic materials, but it also explains the protection and utilization of intellectual property associated with biomedical materials.
Collapse
Affiliation(s)
- Dipankar Das
- Department of Chemical and Biomolecular Engineering, Seoul National University of Science and Technology, Seoul, South Korea.,Convergence Institute of Biomedical Engineering and Biomaterials, Seoul National University of Science and Technology, Seoul, South Korea
| | - Insup Noh
- Department of Chemical and Biomolecular Engineering, Seoul National University of Science and Technology, Seoul, South Korea. .,Convergence Institute of Biomedical Engineering and Biomaterials, Seoul National University of Science and Technology, Seoul, South Korea.
| |
Collapse
|
28
|
Fenton OS, Paolini M, Andresen JL, Müller FJ, Langer R. Outlooks on Three-Dimensional Printing for Ocular Biomaterials Research. J Ocul Pharmacol Ther 2019; 36:7-17. [PMID: 31211652 PMCID: PMC6985767 DOI: 10.1089/jop.2018.0142] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/07/2019] [Indexed: 10/26/2022] Open
Abstract
Given its potential for high-resolution, customizable, and waste-free fabrication of medical devices and in vitro biological models, 3-dimensional (3D) bioprinting has broad utility within the biomaterials field. Indeed, 3D bioprinting has to date been successfully used for the development of drug delivery systems, the recapitulation of hard biological tissues, and the fabrication of cellularized organ and tissue-mimics, among other applications. In this study, we highlight convergent efforts within engineering, cell biology, soft matter, and chemistry in an overview of the 3D bioprinting field, and we then conclude our work with outlooks toward the application of 3D bioprinting for ocular research in vitro and in vivo.
Collapse
Affiliation(s)
- Owen S. Fenton
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Marion Paolini
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Jason L. Andresen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Florence J. Müller
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Robert Langer
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| |
Collapse
|
29
|
Steier A, Muñiz A, Neale D, Lahann J. Emerging Trends in Information-Driven Engineering of Complex Biological Systems. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1806898. [PMID: 30957921 DOI: 10.1002/adma.201806898] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 12/03/2018] [Indexed: 06/09/2023]
Abstract
Synthetic biological systems are used for a myriad of applications, including tissue engineered constructs for in vivo use and microengineered devices for in vitro testing. Recent advances in engineering complex biological systems have been fueled by opportunities arising from the combination of bioinspired materials with biological and computational tools. Driven by the availability of large datasets in the "omics" era of biology, the design of the next generation of tissue equivalents will have to integrate information from single-cell behavior to whole organ architecture. Herein, recent trends in combining multiscale processes to enable the design of the next generation of biomaterials are discussed. Any successful microprocessing pipeline must be able to integrate hierarchical sets of information to capture key aspects of functional tissue equivalents. Micro- and biofabrication techniques that facilitate hierarchical control as well as emerging polymer candidates used in these technologies are also reviewed.
Collapse
Affiliation(s)
- Anke Steier
- Institute of Functional Interfaces (IFG), Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Ayşe Muñiz
- Biointerfaces Institute and Macromolecular Science and Engineering Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Dylan Neale
- Biointerfaces Institute and Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Joerg Lahann
- Institute of Functional Interfaces (IFG), Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
- Biointerfaces Institute, Departments of Chemical Engineering, Materials Science and Engineering, and Biomedical Engineering and the, Macromolecular Science and Engineering Program, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
30
|
Engineering nanocellulose hydrogels for biomedical applications. Adv Colloid Interface Sci 2019; 267:47-61. [PMID: 30884359 DOI: 10.1016/j.cis.2019.03.002] [Citation(s) in RCA: 198] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/06/2019] [Accepted: 03/06/2019] [Indexed: 12/11/2022]
Abstract
Nanocellulose hydrogels are highly hydrated porous cellulosic soft materials with good mechanical properties. These cellulose-based gels can be produced from bacterial or plant cellulose nanofibrils, which are hydrophilic, renewable, biodegradable and biocompatible. Nanocellulose, whether fibrils (CNF), crystals (CNC) or bacterial (BNC), has a high aspect ratio and surface area, and can be chemically modified with functional groups or by grafting biomolecules. Cellulose functionalization provides enhanced physical and chemical properties and control of biological interactions, tailoring its hydrogels for specific applications. Here, we critically review nanocellulose hydrogels for biomedical applications. Nanocellulose hydrogels have been demonstrated for 3D cell culture, mimicking the extracellular matrix (ECM) properties with low cytotoxicity. For wound dressing and cartilage repair, nanocellulose gels promote cell regeneration while providing the required mechanical properties for tissue engineering scaffolds. The encapsulation of therapeutics within nanocellulose allows the targeted delivery of drugs. Currently, cellulose crosslinking to peptides and proteins enables a new generation of low cost and renewable smart materials used in diagnostics. Last, the organized mesh of fibres contained in hydrogels drives applications in separation of biomolecules and cells. Nanocellulose hydrogels have emerged as a highly engineerable platform for multiple biomedical applications, providing renewable and performant solutions to life sciences.
Collapse
|
31
|
|
32
|
Koons GL, Mikos AG. Progress in three-dimensional printing with growth factors. J Control Release 2019; 295:50-59. [PMID: 30579982 PMCID: PMC6358495 DOI: 10.1016/j.jconrel.2018.12.035] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 11/06/2018] [Accepted: 12/19/2018] [Indexed: 12/19/2022]
Abstract
Incorporation of growth factors in biomedical constructs can encourage cellular activities necessary for tissue regeneration within an implant system. Three-dimensional printing offers a capacity for spatial dictation and dosage control of incorporated growth factors which promises to minimize complications from the supraphysiologic doses and burst release involved in current growth factor delivery systems. Successful implementation of three-dimensional printing with growth factors requires preservation of the bioactivity of printed growth factors, spatial localization of growth factors within the construct architecture during printing, and controlled release of growth factors after printing. This review describes demonstrated approaches for addressing each of these goals, including direct inclusion of growth factors with the biomaterial during printing, or intermediary encapsulation of growth factors in delivery vehicles such as microparticles or nanoparticles.
Collapse
Affiliation(s)
- Gerry L Koons
- Department of Bioengineering, Rice University, Houston, TX, USA; Center for Engineering Complex Tissues, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA.
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, TX, USA; Center for Engineering Complex Tissues, USA.
| |
Collapse
|
33
|
Bailey KE, Floren ML, D'Ovidio TJ, Lammers SR, Stenmark KR, Magin CM. Tissue-informed engineering strategies for modeling human pulmonary diseases. Am J Physiol Lung Cell Mol Physiol 2019; 316:L303-L320. [PMID: 30461289 PMCID: PMC6397349 DOI: 10.1152/ajplung.00353.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 11/13/2018] [Accepted: 11/14/2018] [Indexed: 12/14/2022] Open
Abstract
Chronic pulmonary diseases, including idiopathic pulmonary fibrosis (IPF), pulmonary hypertension (PH), and chronic obstructive pulmonary disease (COPD), account for staggering morbidity and mortality worldwide but have limited clinical management options available. Although great progress has been made to elucidate the cellular and molecular pathways underlying these diseases, there remains a significant disparity between basic research endeavors and clinical outcomes. This discrepancy is due in part to the failure of many current disease models to recapitulate the dynamic changes that occur during pathogenesis in vivo. As a result, pulmonary medicine has recently experienced a rapid expansion in the application of engineering principles to characterize changes in human tissues in vivo and model the resulting pathogenic alterations in vitro. We envision that engineering strategies using precision biomaterials and advanced biomanufacturing will revolutionize current approaches to disease modeling and accelerate the development and validation of personalized therapies. This review highlights how advances in lung tissue characterization reveal dynamic changes in the structure, mechanics, and composition of the extracellular matrix in chronic pulmonary diseases and how this information paves the way for tissue-informed engineering of more organotypic models of human pathology. Current translational challenges are discussed as well as opportunities to overcome these barriers with precision biomaterial design and advanced biomanufacturing techniques that embody the principles of personalized medicine to facilitate the rapid development of novel therapeutics for this devastating group of chronic diseases.
Collapse
Affiliation(s)
- Kolene E Bailey
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Michael L Floren
- Cardiovascular Pulmonary Research Laboratories, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
- Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
- Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Tyler J D'Ovidio
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Steven R Lammers
- Department of Bioengineering, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Kurt R Stenmark
- Cardiovascular Pulmonary Research Laboratories, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
- Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
- Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Chelsea M Magin
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
- Department of Bioengineering, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
34
|
Identification of Key Signaling Pathways Orchestrating Substrate Topography Directed Osteogenic Differentiation Through High-Throughput siRNA Screening. Sci Rep 2019; 9:1001. [PMID: 30700820 PMCID: PMC6353928 DOI: 10.1038/s41598-018-37554-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 12/04/2018] [Indexed: 12/14/2022] Open
Abstract
Fibrous scaffolds are used for bone tissue engineering purposes with great success across a variety of polymers with different physical and chemical properties. It is now evident that the correct degree of curvature promotes increased cytoskeletal tension on osteoprogenitors leading to osteogenic differentiation. However, the mechanotransductive pathways involved in this phenomenon are not fully understood. To achieve a reproducible and specific cellular response, an increased mechanistic understanding of the molecular mechanisms driving the fibrous scaffold mediated bone regeneration must be understood. High throughput siRNA mediated screening technology has been utilized for dissecting molecular targets that are important in certain cellular phenotypes. In this study, we used siRNA mediated gene silencing to understand the osteogenic differentiation observed on fibrous scaffolds. A high-throughput siRNA screen was conducted using a library collection of 863 genes including important human kinase and phosphatase targets on pre-osteoblast SaOS-2 cells. The cells were grown on electrospun poly(methyl methacrylate) (PMMA) scaffolds with a diameter of 0.938 ± 0.304 µm and a flat surface control. The osteogenic transcription factor RUNX2 was quantified with an in-cell western (ICW) assay for the primary screen and significant targets were selected via two sample t-test. After selecting the significant targets, a secondary screen was performed to identify osteoinductive markers that also effect cell shape on fibrous topography. Finally, we report the most physiologically relevant molecular signaling mechanisms that are involved in growth factor free, fibrous topography mediated osteoinduction. We identified GTPases, membrane channel proteins, and microtubule associated targets that promote an osteoinductive cell shape on fibrous scaffolds.
Collapse
|
35
|
Dasgupta Q, Madras G, Chatterjee K. Gradient platform for combinatorial screening of thermoset polymers for biomedical applications. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 94:766-777. [PMID: 30423763 DOI: 10.1016/j.msec.2018.10.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 09/17/2018] [Accepted: 10/03/2018] [Indexed: 11/19/2022]
Abstract
The goal of this work was to design a device for rapid screening of crosslinked thermoset polymers. This gradient curing platform is capable of yielding a library of polyesters with systematically varying mechanical and physicochemical properties and the resultant cellular response. A library of poly(xylitolsebacate) polyesters was prepared in this device by differential curing to yield a gradient polymer. The resultant polymer exhibits a gradient in the storage modulus (1 to 5 MPa), wettability (70° < water contact angle < 110°), degree of crosslinking, degradation rate (3-25% in 7 days), drug release and biological response (ability to support stem cell proliferation and differentiation) from one end of the polymer to the other. Primary human mesenchymal stem cells were cultured to assess the cellular response in vitro. Maximal stem cell proliferation and osteogenesis was observed on the highly crosslinked polyester segments that provide high stiffness, are hydrophobic and are slow degrading as compared to the lower cured counterparts. Under in vivo conditions, this material showed differential response across the gradient without displaying significant concerns for inflammation or infection. This gradient curing device is capable of ascertaining suitable curing conditions to obtain appropriate polymers for application specific requirements. This gradient platform was further used to identify optimal processing parameters to prepare three-dimensional tissue scaffolds such as electrospun fiber mats and porous foams. Thus, this versatile combinatorial platform is well suited for rapid screening of thermoset polymers for biomedical applications.
Collapse
Affiliation(s)
- Queeny Dasgupta
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Giridhar Madras
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India; Department of Chemical Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Kaushik Chatterjee
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India; Department of Materials Engineering, Indian Institute of Science, Bangalore 560012, India.
| |
Collapse
|
36
|
|
37
|
Ma Y, Xie L, Yang B, Tian W. Three-dimensional printing biotechnology for the regeneration of the tooth and tooth-supporting tissues. Biotechnol Bioeng 2018; 116:452-468. [PMID: 30475386 DOI: 10.1002/bit.26882] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 11/09/2018] [Accepted: 11/21/2018] [Indexed: 02/05/2023]
Abstract
The tooth and its supporting tissues are organized with complex three-dimensional (3D) architecture, including the dental pulp with a blood supply and nerve tissues, complex multilayer periodontium, and highly aligned periodontal ligament (PDL). Mimicking such 3D complexity and the multicellular interactions naturally existing in dental structures represents great challenges in dental regeneration. Attempts to construct the complex system of the tooth and tooth-supporting apparatus (i.e., the PDL, alveolar bone, and cementum) have made certain progress owing to 3D printing biotechnology. Recent advances have enabled the 3D printing of biocompatible materials, seed cells, and supporting components into complex 3D functional living tissue. Furthermore, 3D bioprinting is driving major innovations in regenerative medicine, giving the field of regenerative dentistry a boost. The fabrication of scaffolds via 3D printing is already being performed extensively at the laboratory bench and in clinical trials; however, printing living cells and matrix materials together to produce tissue constructs by 3D bioprinting remains limited to the regeneration of dental pulp and the tooth germ. This review summarizes the application of scaffolds for cell seeding and biofabricated tissues via 3D printing and bioprinting, respectively, in the tooth and its supporting tissues. Additionally, the key advantages and prospects of 3D bioprinting in regenerative dentistry are highlighted, providing new ideas for dental regeneration.
Collapse
Affiliation(s)
- Yue Ma
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, People's Republic of China.,National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, People's Republic of China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Li Xie
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, People's Republic of China.,National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Bo Yang
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, People's Republic of China.,National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, People's Republic of China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Weidong Tian
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, People's Republic of China.,National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, People's Republic of China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
38
|
Lim HJ, Khan Z, Lu X, Perera TH, Wilems TS, Ravivarapu KT, Smith Callahan LA. Mechanical stabilization of proteolytically degradable polyethylene glycol dimethacrylate hydrogels through peptide interaction. Acta Biomater 2018. [PMID: 29526829 DOI: 10.1016/j.actbio.2018.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Balancing enhancement of neurite extension against loss of matrix support in synthetic hydrogels containing proteolytically degradable and bioactive signaling peptides to optimize tissue formation is difficult. Using a systematic approach, polyethylene glycol hydrogels containing concurrent continuous concentration gradients of the laminin derived bioactive signaling peptide, Ile-Lys-Val-Ala-Val (IKVAV), and collagen derived matrix metalloprotease degradable peptide, GPQGIWGQ, were fabricated and characterized. During proteolytic degradation of the concentration gradient hydrogels, the IKVAV and IWGQ cleavage fragment from GPQGIWGQ were found to interact and stabilize the bulk Young's Modulus of the hydrogel. Further testing of discrete samples containing GPQGIWGQ or its cleavage fragments, GPQG and IWGQ, indicates hydrophobic interactions between the peptides are not necessary for mechanical stabilization of the hydrogel, but changes in the concentration ratio between the peptides tethered in the hydrogel and salts and ions in the swelling solution can affect the stabilization. Encapsulation of human induced pluripotent stem cell derived neural stem cells did not reduce the mechanical properties of the hydrogel over a 14 day neural differentiation culture period, and IKVAV was found to maintain concentration dependent effects on neurite extension and mRNA gene expression of neural cytoskeletal markers, similar to previous studies. As a result, this work has significant implications for the analysis of biological studies in matrices, as the material and mechanical properties of the hydrogel may be unexpectedly temporally changing during culture due to interactions between peptide signaling elements, underscoring the need for greater matrix characterization during the degradation and cell culture. STATEMENT OF SIGNIFICANCE Greater emulation of the native extracellular matrix is necessary for tissue formation. To achieve this, matrices are becoming more complex, often including multiple bioactive signaling elements. However, peptide signaling in polyethylene glycol matrices and amino acids interactions between peptides can affect hydrogel material and mechanical properties, but are rarely studied. The current study identifies such an interaction between laminin derived peptide, IKVAV, and collagen derived matrix metalloprotease degradable peptide, GPQGIWGQ. Previous studies using these peptides did not identify their interactions' ability to mechanically stabilize the hydrogel during degradation. This work underscores the need for greater matrix characterization and consideration of bioactive signaling element effects temporally on the matrix's material and mechanical properties, as they can contribute to cellular response.
Collapse
Affiliation(s)
- Hyun Ju Lim
- The Vivian L Smith Department of Neurosurgery, McGovern Medical School at the University of Texas Health Science Center at Houston, United States; Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School at the University of Texas Health Science Center at Houston, United States
| | - Zara Khan
- The Vivian L Smith Department of Neurosurgery, McGovern Medical School at the University of Texas Health Science Center at Houston, United States; Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School at the University of Texas Health Science Center at Houston, United States
| | - Xi Lu
- The Vivian L Smith Department of Neurosurgery, McGovern Medical School at the University of Texas Health Science Center at Houston, United States; Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School at the University of Texas Health Science Center at Houston, United States
| | - T Hiran Perera
- The Vivian L Smith Department of Neurosurgery, McGovern Medical School at the University of Texas Health Science Center at Houston, United States; Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School at the University of Texas Health Science Center at Houston, United States
| | - Thomas S Wilems
- The Vivian L Smith Department of Neurosurgery, McGovern Medical School at the University of Texas Health Science Center at Houston, United States; Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School at the University of Texas Health Science Center at Houston, United States
| | - Krishna T Ravivarapu
- The Vivian L Smith Department of Neurosurgery, McGovern Medical School at the University of Texas Health Science Center at Houston, United States; Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School at the University of Texas Health Science Center at Houston, United States
| | - Laura A Smith Callahan
- The Vivian L Smith Department of Neurosurgery, McGovern Medical School at the University of Texas Health Science Center at Houston, United States; Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School at the University of Texas Health Science Center at Houston, United States; The Department of Nanomedicine and Biomedical Engineering, McGovern Medical School at the University of Texas Health Science Center at Houston, United States; The MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, United States.
| |
Collapse
|
39
|
Othman Z, Cillero Pastor B, van Rijt S, Habibovic P. Understanding interactions between biomaterials and biological systems using proteomics. Biomaterials 2018; 167:191-204. [PMID: 29571054 DOI: 10.1016/j.biomaterials.2018.03.020] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 03/02/2018] [Accepted: 03/12/2018] [Indexed: 12/22/2022]
Abstract
The role that biomaterials play in the clinical treatment of damaged organs and tissues is changing. While biomaterials used in permanent medical devices were required to passively take over the function of a damaged tissue in the long term, current biomaterials are expected to trigger and harness the self-regenerative potential of the body in situ and then to degrade, the foundation of regenerative medicine. To meet these different requirements, it is imperative to fully understand the interactions biomaterials have with biological systems, in space and in time. This knowledge will lead to a better understanding of the regenerative capabilities of biomaterials aiding their design with improved functionalities (e.g. biocompatibility, bioactivity). Proteins play a pivotal role in the interaction between biomaterials and cells or tissues. Protein adsorption on the material surface is the very first event of this interaction, which is determinant for the subsequent processes of cell growth, differentiation, and extracellular matrix formation. Against this background, the aim of the current review is to provide insight in the current knowledge of the role of proteins in cell-biomaterial and tissue-biomaterial interactions. In particular, the focus is on proteomics studies, mainly using mass spectrometry, and the knowledge they have generated on protein adsorption of biomaterials, protein production by cells cultured on materials, safety and efficacy of new materials based on nanoparticles and the analysis of extracellular matrices and extracellular matrix-derived products. In the outlook, the potential and limitations of this approach are discussed and mass spectrometry imaging is presented as a powerful technique that complements existing mass spectrometry techniques by providing spatial molecular information about the material-biological system interactions.
Collapse
Affiliation(s)
- Ziryan Othman
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Instructive Biomaterials Engineering, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Berta Cillero Pastor
- The Maastricht Multimodal Molecular Imaging Institute (M4I), Division of Imaging Mass Spectrometry, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Sabine van Rijt
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Instructive Biomaterials Engineering, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Pamela Habibovic
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Instructive Biomaterials Engineering, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands.
| |
Collapse
|
40
|
Smith Callahan LA. Gradient Material Strategies for Hydrogel Optimization in Tissue Engineering Applications. High Throughput 2018; 7:E1. [PMID: 29485612 PMCID: PMC5876527 DOI: 10.3390/ht7010001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 12/30/2017] [Accepted: 01/02/2018] [Indexed: 12/15/2022] Open
Abstract
Although a number of combinatorial/high-throughput approaches have been developed for biomaterial hydrogel optimization, a gradient sample approach is particularly well suited to identify hydrogel property thresholds that alter cellular behavior in response to interacting with the hydrogel due to reduced variation in material preparation and the ability to screen biological response over a range instead of discrete samples each containing only one condition. This review highlights recent work on cell-hydrogel interactions using a gradient material sample approach. Fabrication strategies for composition, material and mechanical property, and bioactive signaling gradient hydrogels that can be used to examine cell-hydrogel interactions will be discussed. The effects of gradients in hydrogel samples on cellular adhesion, migration, proliferation, and differentiation will then be examined, providing an assessment of the current state of the field and the potential of wider use of the gradient sample approach to accelerate our understanding of matrices on cellular behavior.
Collapse
Affiliation(s)
- Laura A Smith Callahan
- The Vivian L. Smith Department of Neurosurgery, Center for Stem Cell & Regenerative Medicine, and Department of Nanomedicine and Biomedical Engineering, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
41
|
Maffioli E, Schulte C, Nonnis S, Grassi Scalvini F, Piazzoni C, Lenardi C, Negri A, Milani P, Tedeschi G. Proteomic Dissection of Nanotopography-Sensitive Mechanotransductive Signaling Hubs that Foster Neuronal Differentiation in PC12 Cells. Front Cell Neurosci 2018; 11:417. [PMID: 29354032 PMCID: PMC5758595 DOI: 10.3389/fncel.2017.00417] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/12/2017] [Indexed: 12/11/2022] Open
Abstract
Neuronal cells are competent in precisely sensing nanotopographical features of their microenvironment. The perceived microenvironmental information will be “interpreted” by mechanotransductive processes and impacts on neuronal functioning and differentiation. Attempts to influence neuronal differentiation by engineering substrates that mimic appropriate extracellular matrix (ECM) topographies are hampered by the fact that profound details of mechanosensing/-transduction complexity remain elusive. Introducing omics methods into these biomaterial approaches has the potential to provide a deeper insight into the molecular processes and signaling cascades underlying mechanosensing/-transduction but their exigence in cellular material is often opposed by technical limitations of major substrate top-down fabrication methods. Supersonic cluster beam deposition (SCBD) allows instead the bottom-up fabrication of nanostructured substrates over large areas characterized by a quantitatively controllable ECM-like nanoroughness that has been recently shown to foster neuron differentiation and maturation. Exploiting this capacity of SCBD, we challenged mechanosensing/-transduction and differentiative behavior of neuron-like PC12 cells with diverse nanotopographies and/or changes of their biomechanical status, and analyzed their phosphoproteomic profiles in these settings. Versatile proteins that can be associated to significant processes along the mechanotransductive signal sequence, i.e., cell/cell interaction, glycocalyx and ECM, membrane/f-actin linkage and integrin activation, cell/substrate interaction, integrin adhesion complex, actomyosin organization/cellular mechanics, nuclear organization, and transcriptional regulation, were affected. The phosphoproteomic data suggested furthermore an involvement of ILK, mTOR, Wnt, and calcium signaling in these nanotopography- and/or cell mechanics-related processes. Altogether, potential nanotopography-sensitive mechanotransductive signaling hubs participating in neuronal differentiation were dissected.
Collapse
Affiliation(s)
- Elisa Maffioli
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy
| | - Carsten Schulte
- Centre for Nanostructured Materials and Interfaces, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| | - Simona Nonnis
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| | - Francesca Grassi Scalvini
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| | - Claudio Piazzoni
- Centre for Nanostructured Materials and Interfaces, Università degli Studi di Milano, Milan, Italy
| | - Cristina Lenardi
- Centre for Nanostructured Materials and Interfaces, Università degli Studi di Milano, Milan, Italy
| | - Armando Negri
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| | - Paolo Milani
- Centre for Nanostructured Materials and Interfaces, Università degli Studi di Milano, Milan, Italy
| | - Gabriella Tedeschi
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| |
Collapse
|
42
|
Darnell M, Mooney DJ. Leveraging advances in biology to design biomaterials. NATURE MATERIALS 2017; 16:1178-1185. [PMID: 29170558 DOI: 10.1038/nmat4991] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 08/25/2017] [Indexed: 05/06/2023]
Abstract
Biomaterials have dramatically increased in functionality and complexity, allowing unprecedented control over the cells that interact with them. From these engineering advances arises the prospect of improved biomaterial-based therapies, yet practical constraints favour simplicity. Tools from the biology community are enabling high-resolution and high-throughput bioassays that, if incorporated into a biomaterial design framework, could help achieve unprecedented functionality while minimizing the complexity of designs by identifying the most important material parameters and biological outputs. However, to avoid data explosions and to effectively match the information content of an assay with the goal of the experiment, material screens and bioassays must be arranged in specific ways. By borrowing methods to design experiments and workflows from the bioprocess engineering community, we outline a framework for the incorporation of next-generation bioassays into biomaterials design to effectively optimize function while minimizing complexity. This framework can inspire biomaterials designs that maximize functionality and translatability.
Collapse
Affiliation(s)
- Max Darnell
- Harvard School of Engineering and Applied Sciences, Cambridge, Massachusetts 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Cambridge, Massachusetts 02138, USA
| | - David J Mooney
- Harvard School of Engineering and Applied Sciences, Cambridge, Massachusetts 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Cambridge, Massachusetts 02138, USA
| |
Collapse
|
43
|
Huang G, Li F, Zhao X, Ma Y, Li Y, Lin M, Jin G, Lu TJ, Genin GM, Xu F. Functional and Biomimetic Materials for Engineering of the Three-Dimensional Cell Microenvironment. Chem Rev 2017; 117:12764-12850. [PMID: 28991456 PMCID: PMC6494624 DOI: 10.1021/acs.chemrev.7b00094] [Citation(s) in RCA: 486] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The cell microenvironment has emerged as a key determinant of cell behavior and function in development, physiology, and pathophysiology. The extracellular matrix (ECM) within the cell microenvironment serves not only as a structural foundation for cells but also as a source of three-dimensional (3D) biochemical and biophysical cues that trigger and regulate cell behaviors. Increasing evidence suggests that the 3D character of the microenvironment is required for development of many critical cell responses observed in vivo, fueling a surge in the development of functional and biomimetic materials for engineering the 3D cell microenvironment. Progress in the design of such materials has improved control of cell behaviors in 3D and advanced the fields of tissue regeneration, in vitro tissue models, large-scale cell differentiation, immunotherapy, and gene therapy. However, the field is still in its infancy, and discoveries about the nature of cell-microenvironment interactions continue to overturn much early progress in the field. Key challenges continue to be dissecting the roles of chemistry, structure, mechanics, and electrophysiology in the cell microenvironment, and understanding and harnessing the roles of periodicity and drift in these factors. This review encapsulates where recent advances appear to leave the ever-shifting state of the art, and it highlights areas in which substantial potential and uncertainty remain.
Collapse
Affiliation(s)
- Guoyou Huang
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Fei Li
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
- Department of Chemistry, School of Science,
Xi’an Jiaotong University, Xi’an 710049, People’s Republic
of China
| | - Xin Zhao
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
- Interdisciplinary Division of Biomedical
Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong,
People’s Republic of China
| | - Yufei Ma
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Yuhui Li
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Min Lin
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Guorui Jin
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Tian Jian Lu
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
- MOE Key Laboratory for Multifunctional Materials
and Structures, Xi’an Jiaotong University, Xi’an 710049,
People’s Republic of China
| | - Guy M. Genin
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
- Department of Mechanical Engineering &
Materials Science, Washington University in St. Louis, St. Louis 63130, MO,
USA
- NSF Science and Technology Center for
Engineering MechanoBiology, Washington University in St. Louis, St. Louis 63130,
MO, USA
| | - Feng Xu
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| |
Collapse
|
44
|
Ji S, Guvendiren M. Recent Advances in Bioink Design for 3D Bioprinting of Tissues and Organs. Front Bioeng Biotechnol 2017; 5:23. [PMID: 28424770 PMCID: PMC5380738 DOI: 10.3389/fbioe.2017.00023] [Citation(s) in RCA: 221] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/21/2017] [Indexed: 12/26/2022] Open
Abstract
There is a growing demand for alternative fabrication approaches to develop tissues and organs as conventional techniques are not capable of fabricating constructs with required structural, mechanical, and biological complexity. 3D bioprinting offers great potential to fabricate highly complex constructs with precise control of structure, mechanics, and biological matter [i.e., cells and extracellular matrix (ECM) components]. 3D bioprinting is an additive manufacturing approach that utilizes a "bioink" to fabricate devices and scaffolds in a layer-by-layer manner. 3D bioprinting allows printing of a cell suspension into a tissue construct with or without a scaffold support. The most common bioinks are cell-laden hydrogels, decellulerized ECM-based solutions, and cell suspensions. In this mini review, a brief description and comparison of the bioprinting methods, including extrusion-based, droplet-based, and laser-based bioprinting, with particular focus on bioink design requirements are presented. We also present the current state of the art in bioink design including the challenges and future directions.
Collapse
Affiliation(s)
- Shen Ji
- Instructive Biomaterials and Additive Manufacturing (IBAM) Laboratory, Otto H. York Department of Chemical Biological and Pharmaceutical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Murat Guvendiren
- Instructive Biomaterials and Additive Manufacturing (IBAM) Laboratory, Otto H. York Department of Chemical Biological and Pharmaceutical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| |
Collapse
|