1
|
Semitela A, Marques PAAP, Completo A. Strategies to engineer articular cartilage with biomimetic zonal features: a review. Biomater Sci 2024. [PMID: 39463257 DOI: 10.1039/d4bm00579a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Articular cartilage (AC) is a highly specialized tissue with restricted ability for self-regeneration, given its avascular and acellular nature. Although a considerable number of surgical treatments is available for the repair, reconstruction, and regeneration of AC defects, most of them do not prioritize the development of engineered cartilage with zonal stratification derived from biomimetic biochemical, biomechanical and topographic cues. In the absence of these zonal elements, engineered cartilage will exhibit increased susceptibility to failure and will neither be able to withstand the mechanical loading to which AC is subjected nor will it integrate well with the surrounding tissue. In this regard, new breakthroughs in the development of hierarchical stratified engineered cartilage are highly sought after. Initially, this review provides a comprehensive analysis of the composition and zonal organization of AC, aiming to enhance our understanding of the significance of the structure of AC for its function. Next, we direct our attention towards the existing in vitro and in vivo studies that introduce zonal elements in engineered cartilage to elicit appropriate AC regeneration by employing tissue engineering strategies. Finally, the advantages, challenges, and future perspectives of these approaches are presented.
Collapse
Affiliation(s)
- Angela Semitela
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Paula A A P Marques
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - António Completo
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal.
| |
Collapse
|
2
|
Fernández-Galiana Á, Bibikova O, Vilms Pedersen S, Stevens MM. Fundamentals and Applications of Raman-Based Techniques for the Design and Development of Active Biomedical Materials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2210807. [PMID: 37001970 DOI: 10.1002/adma.202210807] [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: 11/21/2022] [Revised: 03/03/2023] [Indexed: 06/19/2023]
Abstract
Raman spectroscopy is an analytical method based on light-matter interactions that can interrogate the vibrational modes of matter and provide representative molecular fingerprints. Mediated by its label-free, non-invasive nature, and high molecular specificity, Raman-based techniques have become ubiquitous tools for in situ characterization of materials. This review comprehensively describes the theoretical and practical background of Raman spectroscopy and its advanced variants. The numerous facets of material characterization that Raman scattering can reveal, including biomolecular identification, solid-to-solid phase transitions, and spatial mapping of biomolecular species in bioactive materials, are highlighted. The review illustrates the potential of these techniques in the context of active biomedical material design and development by highlighting representative studies from the literature. These studies cover the use of Raman spectroscopy for the characterization of both natural and synthetic biomaterials, including engineered tissue constructs, biopolymer systems, ceramics, and nanoparticle formulations, among others. To increase the accessibility and adoption of these techniques, the present review also provides the reader with practical recommendations on the integration of Raman techniques into the experimental laboratory toolbox. Finally, perspectives on how recent developments in plasmon- and coherently-enhanced Raman spectroscopy can propel Raman from underutilized to critical for biomaterial development are provided.
Collapse
Affiliation(s)
- Álvaro Fernández-Galiana
- Department of Materials, Department of Bioengineering, Imperial College London, SW7 2AZ, London, UK
| | - Olga Bibikova
- Department of Materials, Department of Bioengineering, Imperial College London, SW7 2AZ, London, UK
| | - Simon Vilms Pedersen
- Department of Materials, Department of Bioengineering, Imperial College London, SW7 2AZ, London, UK
| | - Molly M Stevens
- Department of Materials, Department of Bioengineering, Imperial College London, SW7 2AZ, London, UK
| |
Collapse
|
3
|
Raja Somu D, Fuentes M, Lou L, Agarwal A, Porter M, Merk V. Revealing chemistry-structure-function relationships in shark vertebrae across length scales. Acta Biomater 2024:S1742-7061(24)00567-1. [PMID: 39349113 DOI: 10.1016/j.actbio.2024.09.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/22/2024] [Accepted: 09/24/2024] [Indexed: 10/02/2024]
Abstract
Shark cartilage presents a complex material composed of collagen, proteoglycans, and bioapatite. In the present study, we explored the link between microstructure, chemical composition, and biomechanical function of shark vertebral cartilage using Polarized Light Microscopy (PLM), Atomic Force Microscopy (AFM), Confocal Raman Microspectroscopy, and Nanoindentation. Our investigation focused on vertebrae from Blacktip and Shortfin Mako sharks. As typical representatives of the orders Carcharhiniformes and Lamniformes, these species differ in preferred habitat, ecological role, and swimming style. We observed structural variations in mineral organization and collagen fiber arrangement using PLM and AFM. In both sharks, the highly calcified corpus calcarea shows a ridged morphology, while a chain-like network is present in the less mineralized intermedialia. Raman spectromicroscopy demonstrates a relative increase of glucosaminocycans (GAGs) with respect to collagen and a decrease in mineral-rich zones, underlining the role of GAGs in modulating bioapatite mineralization. Region-specific testing confirmed that intravertebral variations in mineral content and arrangement result in distinct nanomechanical properties. Local Young's moduli from mineralized regions exceeded bulk values by a factor of 10. Overall, this work provides profound insights into a flexible yet strong biocomposite, which is crucial for the extraordinary speed of cartilaginous fish in the worlds' oceans. STATEMENT OF SIGNIFICANCE: Shark cartilage is a morphologically complex material composed of collagen, sulfated proteoglycans, and calcium phosphate minerals. This study explores the link between microstructure, chemical composition, and biological mechanical function of shark vertebral cartilage at the micro- and nanometer scale in typical Carcharhiniform and Lamniform shark species, which represent different vertebral mineralization morphologies, swimming styles and speeds. By studying the intricacies of shark vertebrae, we hope to lay the foundation for biomimetic composite materials that harness lamellar reinforcement and tailored stiffness gradients, capable of dynamic and localized adjustments during movement.
Collapse
Affiliation(s)
- Dawn Raja Somu
- Department of Chemistry and Biochemistry, Department of Ocean and Mechanical Engineering, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Malena Fuentes
- Department of Chemistry and Biochemistry, Department of Ocean and Mechanical Engineering, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Lihua Lou
- Department of Mechanical and Materials Engineering, Florida International University, Miami, FL 33174, USA
| | - Arvind Agarwal
- Department of Mechanical and Materials Engineering, Florida International University, Miami, FL 33174, USA
| | - Marianne Porter
- Department of Biological Sciences, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Vivian Merk
- Department of Chemistry and Biochemistry, Department of Ocean and Mechanical Engineering, Florida Atlantic University, Boca Raton, FL 33431, USA.
| |
Collapse
|
4
|
Barthold JE, Cai L, McCreery KP, Fischenich KM, Eckstein KN, Ferguson VL, Emery NC, Breur G, Neu CP. Integrative cartilage repair using acellular allografts for engineered structure and surface lubrication in vivo. NPJ Regen Med 2024; 9:25. [PMID: 39341829 PMCID: PMC11438864 DOI: 10.1038/s41536-024-00367-x] [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: 12/04/2022] [Accepted: 09/10/2024] [Indexed: 10/01/2024] Open
Abstract
The repair of articular cartilage after damage is challenging, and decellularized tissue offers a possible treatment option to promote regeneration. Here, we show that acellular osteochondral allografts improve integrative cartilage repair compared to untreated defects after 6 months in an ovine model. Functional measures of intratissue strain/structure assessed by MRI demonstrate similar biomechanics of implants and native cartilage. Compared to native tissue and defects, the structure, composition, and tribology of acellular allografts preserve surface roughness and lubrication, material properties under compression and relaxation, compositional ratios of collagen:glycosaminoglycan and collagen:phosphate, and relative composition of types I/II collagen. While high cellularity was observed in bone regions and integration zones between cartilage-allografts, recellularization of chondral implants was inconsistent, with cell migration typically less than ~750 µm into the dense decellularized tissue, possibly limiting long-term cellular maintenance. Our results demonstrate the structural and biomechanical efficacy of acellular allografts for at least six months in vivo.
Collapse
Affiliation(s)
- Jeanne E Barthold
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Luyao Cai
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Kaitlin P McCreery
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO, USA
| | - Kristine M Fischenich
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Kevin N Eckstein
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Virginia L Ferguson
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, USA
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Nancy C Emery
- Department of Ecology and Evolutionary Biology, University of Colorado Boulder, Boulder, CO, USA
| | - Gert Breur
- Department of Veterinary Clinical Services, Purdue University, West Lafayette, IN, USA
| | - Corey P Neu
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, USA.
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA.
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO, USA.
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA.
| |
Collapse
|
5
|
Raj P, Wu L, Almeida C, Conway L, Tanwar S, Middendorf J, Barman I. Shining Light on Osteoarthritis: Spatially Offset Raman Spectroscopy as a Window into Cartilage Health. ACS Sens 2024; 9:3794-3804. [PMID: 38976969 DOI: 10.1021/acssensors.4c01242] [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] [Indexed: 07/10/2024]
Abstract
Articular cartilage is a complex tissue, and early detection of osteoarthritis (OA) is crucial for effective treatment. However, current imaging modalities lack molecular specificity and primarily detect late-stage changes. In this study, we propose the use of spatially offset Raman spectroscopy (SORS) for noninvasive, depth-dependent, and molecular-specific diagnostics of articular cartilage. We demonstrate the potential of SORS to penetrate deep layers of cartilage, providing a comprehensive understanding of disease progression. Our SORS measurements were characterized and validated through mechanical and histological techniques, revealing strong correlations between spectroscopic measurements and both Young's modulus and depth of cartilage damage. By longitudinally monitoring enzymatically degraded condyles, we further developed a depth-dependent damage-tracking method. Our analysis revealed distinct components related to sample depth and glycosaminoglycan (GAG) changes, offering a comprehensive picture of cartilage health. Collectively, these findings highlight the potential of SORS as a valuable tool for enhancing OA management and improving patient outcomes.
Collapse
Affiliation(s)
- Piyush Raj
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Lintong Wu
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Craig Almeida
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Lauren Conway
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Swati Tanwar
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Jill Middendorf
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Ishan Barman
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University, School of Medicine, Baltimore, Maryland 21205, United States
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland 21287, United States
| |
Collapse
|
6
|
Cook H, Crisford A, Bourdakos K, Dunlop D, Oreffo ROC, Mahajan S. Holistic vibrational spectromics assessment of human cartilage for osteoarthritis diagnosis. BIOMEDICAL OPTICS EXPRESS 2024; 15:4264-4280. [PMID: 39022535 PMCID: PMC11249685 DOI: 10.1364/boe.520171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 07/20/2024]
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease, presented as wearing down of articular cartilage and resulting in pain and limited mobility for 1 in 10 adults in the UK [Osteoarthr. Cartil.28(6), 792 (2020)10.1016/j.joca.2020.03.004]. There is an unmet need for patient friendly paradigms for clinical assessment that do not use ionizing radiation (CT), exogenous contrast enhancing dyes (MRI), and biopsy. Hence, techniques that use non-destructive, near- and shortwave infrared light (NIR, SWIR) may be ideal for providing label-free, deep tissue interrogation. This study demonstrates multimodal "spectromics", low-level abstraction data fusion of non-destructive NIR Raman scattering spectroscopy and NIR-SWIR absorption spectroscopy, providing an enhanced, interpretable "fingerprint" for diagnosis of OA in human cartilage. This is proposed as method level innovation applicable to both arthro- or endoscopic (minimally invasive) or potential exoscopic (non-invasive) optical approaches. Samples were excised from femoral heads post hip arthroplasty from OA patients (n = 13) and age-matched control (osteoporosis) patients (n = 14). Under multivariate statistical analysis and supervised machine learning, tissue was classified to high precision: 100% segregation of tissue classes (using 10 principal components), and a classification accuracy of 95% (control) and 80% (OA), using the combined vibrational data. There was a marked performance improvement (5 to 6-fold for multivariate analysis) using the spectromics fingerprint compared to results obtained from solely Raman or NIR-SWIR data. Furthermore, clinically relevant tissue components were identified through discriminatory spectral features - spectromics biomarkers - allowing interpretable feedback from the enhanced fingerprint. In summary, spectromics provides comprehensive information for early OA detection and disease stratification, imperative for effective intervention in treating the degenerative onset disease for an aging demographic. This novel and elegant approach for data fusion is compatible with various NIR-SWIR optical devices that will allow deep non-destructive penetration.
Collapse
Affiliation(s)
- Hiroki Cook
- School of Chemistry, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Anna Crisford
- School of Chemistry, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- Human Development Health, Faculty of Medicine, Southampton SO16 6YD, UK
| | - Konstantinos Bourdakos
- School of Chemistry, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Douglas Dunlop
- University Hospital Southampton NHS Foundation Trust, Southampton, SO16 6YD, UK
| | - Richard O. C. Oreffo
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- Human Development Health, Faculty of Medicine, Southampton SO16 6YD, UK
| | - Sumeet Mahajan
- School of Chemistry, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- Department of Biotechnology, Inland Norway University of Applied Sciences, N-2317 Hamar, Norway
| |
Collapse
|
7
|
Georgiev D, Pedersen SV, Xie R, Fernández-Galiana Á, Stevens MM, Barahona M. RamanSPy: An Open-Source Python Package for Integrative Raman Spectroscopy Data Analysis. Anal Chem 2024; 96:8492-8500. [PMID: 38747470 PMCID: PMC11140669 DOI: 10.1021/acs.analchem.4c00383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 05/08/2024] [Accepted: 05/08/2024] [Indexed: 05/29/2024]
Abstract
Raman spectroscopy is a nondestructive and label-free chemical analysis technique, which plays a key role in the analysis and discovery cycle of various branches of science. Nonetheless, progress in Raman spectroscopic analysis is still impeded by the lack of software, methodological and data standardization, and the ensuing fragmentation and lack of reproducibility of analysis workflows thereof. To address these issues, we introduce RamanSPy, an open-source Python package for Raman spectroscopic research and analysis. RamanSPy provides a comprehensive library of tools for spectroscopic analysis that supports day-to-day tasks, integrative analyses, the development of methods and protocols, and the integration of advanced data analytics. RamanSPy is modular and open source, not tied to a particular technology or data format, and can be readily interfaced with the burgeoning ecosystem for data science, statistical analysis, and machine learning in Python. RamanSPy is hosted at https://github.com/barahona-research-group/RamanSPy, supplemented with extended online documentation, available at https://ramanspy.readthedocs.io, that includes tutorials, example applications, and details about the real-world research applications presented in this paper.
Collapse
Affiliation(s)
- Dimitar Georgiev
- Department
of Computing & UKRI Centre
for Doctoral Training in AI for Healthcare, Imperial College London, London SW7 2AZ, United
Kingdom
- Department
of Materials, Department of Bioengineering & Institute of Biomedical
Engineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Simon Vilms Pedersen
- Department
of Materials, Department of Bioengineering & Institute of Biomedical
Engineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Ruoxiao Xie
- Department
of Materials, Department of Bioengineering & Institute of Biomedical
Engineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Álvaro Fernández-Galiana
- Department
of Materials, Department of Bioengineering & Institute of Biomedical
Engineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Molly M. Stevens
- Department
of Materials, Department of Bioengineering & Institute of Biomedical
Engineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Mauricio Barahona
- Department
of Mathematics, Imperial College London, London SW7 2AZ, United Kingdom
| |
Collapse
|
8
|
Deng W, He Q, Zhang W. Analysis of the mechanism of curcumin against osteoarthritis using metabolomics and transcriptomics. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3313-3329. [PMID: 37938371 PMCID: PMC11074044 DOI: 10.1007/s00210-023-02785-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/12/2023] [Indexed: 11/09/2023]
Abstract
Curcumin, a polyphenolic compound derived from the turmeric plant (Curcuma longa), has been extensively studied for its anti-inflammatory and anti-proliferative properties. The safety and efficacy of curcumin have been thoroughly validated. Nevertheless, the underlying mechanism for treating osteoarthritis remains ambiguous. This study aims to reveal the potential mechanism of curcumin in treating osteoarthritis by using metabolomics and transcriptomics. Firstly, we validated the effect of curcumin on inflammatory factors in human articular chondrocytes. Secondly, we explored the cellular metabolism mechanism of curcumin against osteoarthritis using cell metabolomics. Thirdly, we assessed the differences in gene expression of human articular chondrocytes through transcriptomics. Lastly, to evaluate the essential targets and elucidate the potential mechanism underlying the therapeutic effects of curcumin in osteoarthritis, we conducted a screening of the proteins within the shared pathway of metabolomics and transcriptomics. Our results demonstrated that curcumin significantly decreased the levels of inflammatory markers, such as IL-β, IL-6, and TNF-α, in human articular chondrocytes. Cell metabolomics identified 106 differential metabolites, including beta-aminopropionitrile, 3-amino-2-piperidone, pyrrole-2-carboxaldehyde, and various other components. The transcriptomic analysis yielded 1050 differential mRNAs. Enrichment analysis showed that the differential metabolites and mRNAs were significantly enriched in seven pathways, including glycine, serine, and threonine metabolism; pentose and glucuronate interconversions; glycerolipid metabolism; histidine metabolism; mucin-type o-glycan biosynthesis; inositol phosphate metabolism; and cysteine and methionine metabolism. A total of 23 key targets were identified to be involved in these pathways. We speculate that curcumin may alleviate osteoarthritis by targeting key proteins involved in glycine, serine, and threonine metabolism; inhibiting pyruvate production; and modulating glycolysis.
Collapse
Affiliation(s)
- Wenxiang Deng
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Qinghu He
- Department of Rehabilitation and Healthcare, Hunan University of Medicine, Huaihua, 418000, Hunan, China.
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| | - Wenan Zhang
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| |
Collapse
|
9
|
Noro J, Vilaça-Faria H, Reis RL, Pirraco RP. Extracellular matrix-derived materials for tissue engineering and regenerative medicine: A journey from isolation to characterization and application. Bioact Mater 2024; 34:494-519. [PMID: 38298755 PMCID: PMC10827697 DOI: 10.1016/j.bioactmat.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/19/2023] [Accepted: 01/03/2024] [Indexed: 02/02/2024] Open
Abstract
Biomaterial choice is an essential step during the development tissue engineering and regenerative medicine (TERM) applications. The selected biomaterial must present properties allowing the physiological-like recapitulation of several processes that lead to the reestablishment of homeostatic tissue or organ function. Biomaterials derived from the extracellular matrix (ECM) present many such properties and their use in the field has been steadily increasing. Considering this growing importance, it becomes imperative to provide a comprehensive overview of ECM biomaterials, encompassing their sourcing, processing, and integration into TERM applications. This review compiles the main strategies used to isolate and process ECM-derived biomaterials as well as different techniques used for its characterization, namely biochemical and chemical, physical, morphological, and biological. Lastly, some of their applications in the TERM field are explored and discussed.
Collapse
Affiliation(s)
- Jennifer Noro
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Helena Vilaça-Faria
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Rui L. Reis
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Rogério P. Pirraco
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga, Guimarães, Portugal
| |
Collapse
|
10
|
Blom RP, Rahim D, Paardekam E, Kerkhoffs GMMJ, Iannuzzi D, Smit TH. A Traumatic Impact Immediately Changes the Mechanical Properties of Articular Cartilage. Cartilage 2024:19476035241235633. [PMID: 38501455 DOI: 10.1177/19476035241235633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/20/2024] Open
Abstract
OBJECTIVE To investigate whether and how a single traumatic impact changes the mechanical properties of talar articular cartilage. DESIGN A marble was placed on the joint surface and a weight was dropped on both medial and lateral caprine talus to create a well-defined single focal impact. The mechanical properties of intact and impacted talar cartilage were measured with a micro-indenter. Elastic (storage) and viscous (loss) moduli were determined by oscillatory ramp and dynamic mechanical analysis protocols. RESULTS We found significant differences between ankles and within the same ankle joint, with the medial talus having significantly higher storage- and loss moduli than the lateral talus. The storage- and loss moduli of intact articular cartilage increased with greater indentation depths. However, postimpact the storage- and loss moduli were significantly and consistently lower in all specimens indicating immediate posttraumatic damage. The deeper regions of talar cartilage were less affected by the impact than the more superficial regions. CONCLUSIONS A single traumatic impact results in an immediate and significant decrease of storage- and loss moduli. Further research must focus on the development of non- or minimally invasive diagnostic tools to address the exact microdamage caused by the impact. We speculate that the traumatic impact damaged the collagen fibers that confine the water-binding proteoglycans and thereby decreasing the hydrostatic pressure of cartilage. As part of the treatment directly after a trauma, one could imagine a reduction or restriction of peak loads to prevent the progression of the cascade towards PTOA of the ankle joint.
Collapse
Affiliation(s)
- Robin P Blom
- Department of Orthopedic Surgery and Sports Medicine, Amsterdam University Medical Centers, Amsterdam UMC location AMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Movement Sciences, Sports, Amsterdam, The Netherlands
- Academic Center for Evidence-based Sports Medicine (ACES), Amsterdam, The Netherlands
- IOC Research Center, Amsterdam Collaboration on Health and Safety in Sports (ACHSS), Amsterdam, The Netherlands
| | - Danka Rahim
- Department of Orthopedic Surgery and Sports Medicine, Amsterdam University Medical Centers, Amsterdam UMC location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Erik Paardekam
- Department of Physics and Astronomy and LaserLaB, VU, Amsterdam, The Netherlands
| | - Gino M M J Kerkhoffs
- Department of Orthopedic Surgery and Sports Medicine, Amsterdam University Medical Centers, Amsterdam UMC location AMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Movement Sciences, Sports, Amsterdam, The Netherlands
- Academic Center for Evidence-based Sports Medicine (ACES), Amsterdam, The Netherlands
- IOC Research Center, Amsterdam Collaboration on Health and Safety in Sports (ACHSS), Amsterdam, The Netherlands
| | - Davide Iannuzzi
- Department of Physics and Astronomy and LaserLaB, VU, Amsterdam, The Netherlands
| | - Theodoor H Smit
- Department of Orthopedic Surgery and Sports Medicine, Amsterdam University Medical Centers, Amsterdam UMC location AMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Movement Sciences, Sports, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Adachi T, Tahara Y, Yamamoto K, Yamamoto T, Kanamura N, Akiyoshi K, Mazda O. Cholesterol-Bearing Polysaccharide-Based Nanogels for Development of Novel Immunotherapy and Regenerative Medicine. Gels 2024; 10:206. [PMID: 38534624 DOI: 10.3390/gels10030206] [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: 12/23/2023] [Revised: 02/26/2024] [Accepted: 03/13/2024] [Indexed: 03/28/2024] Open
Abstract
Novel functional biomaterials are expected to bring about breakthroughs in developing immunotherapy and regenerative medicine through their application as drug delivery systems and scaffolds. Nanogels are defined as nanoparticles with a particle size of 100 nm or less and as having a gel structure. Nanogels have a three-dimensional network structure of cross-linked polymer chains, which have a high water content, a volume phase transition much faster than that of a macrogel, and a quick response to external stimuli. As it is possible to transmit substances according to the three-dimensional mesh size of the gel, a major feature is that relatively large substances, such as proteins and nucleic acids, can be taken into the gel. Furthermore, by organizing nanogels as a building block, they can be applied as a scaffold material for tissue regeneration. This review provides a brief overview of the current developments in nanogels in general, especially drug delivery, therapeutic applications, and tissue engineering. In particular, polysaccharide-based nanogels are interesting because they have excellent complexation properties and are highly biocompatible.
Collapse
Affiliation(s)
- Tetsuya Adachi
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, 465 Kajii-cho, Kyoto 602-8566, Japan
| | - Yoshiro Tahara
- Department of Chemical Engineering and Materials Science, Doshisha University, 1-3 Tatara Miyakodani, Kyoto-fu, Kyotanabe-shi 610-0321, Japan
| | - Kenta Yamamoto
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, 465 Kajii-cho, Kyoto 602-8566, Japan
| | - Toshiro Yamamoto
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Narisato Kanamura
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Kazunari Akiyoshi
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Osam Mazda
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, 465 Kajii-cho, Kyoto 602-8566, Japan
| |
Collapse
|
12
|
Pereira L, Echarte L, Romero M, Grazioli G, Pérez-Campos H, Francia A, Vicentino W, Mombrú AW, Faccio R, Álvarez I, Touriño C, Pardo H. Synthesis and characterization of a bovine collagen: GAG scaffold with Uruguayan raw material for tissue engineering. Cell Tissue Bank 2024; 25:123-142. [PMID: 34536180 DOI: 10.1007/s10561-021-09960-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 09/06/2021] [Indexed: 11/28/2022]
Abstract
Tissue engineering (TE) and regenerative medicine offer strategies to improve damaged tissues by using scaffolds and cells. The use of collagen-based biomaterials in the field of TE has been intensively growing over the past decades. Mesenchymal stromal cells (MSCs) and dental pulp stem cells (DPSCs) are promising cell candidates for development of clinical composites. In this study, we proposed the development of a bovine collagen type I: chondroitin-6-sulphate (CG) scaffold, obtained from Uruguayan raw material (certified as free bovine spongiform encephalopathy), with CG crosslinking enhancement using different gamma radiation doses. Structural, biomechanical and chemical characteristics of the scaffolds were assessed by Scanning Electron Microscopy, axial tensile tests, FT-IR and Raman Spectroscopy, respectively. Once we selected the most appropriate scaffold for future use as a TE product, we studied the behavior of MSCs and DPSCs cultured on the scaffold by cytotoxicity, proliferation and differentiation assays. Among the diverse porous scaffolds obtained, the one with the most adequate properties was the one exposed to 15 kGy of gamma radiation. This radiation dose contributed to the crosslinking of molecules, to the formation of new bonds and/or to the reorganization of the collagen fibers. The selected scaffold was non-cytotoxic for the tested cells and a suitable substrate for cell proliferation. Furthermore, the scaffold allowed MSCs differentiation to osteogenic, chondrogenic, and adipogenic lineages. Thus, this work shows a promising approach to the synthesis of a collagen-scaffold suitable for TE.
Collapse
Affiliation(s)
- L Pereira
- Centro NanoMat, Facultad de Química, Instituto Polo Tecnológico de Pando, UdelaR, Camino Aparicio Saravia s/n, 9100, Pando, Canelones, Uruguay
| | - L Echarte
- Área Terapia Celular y Medicina Regenerativa (ATCMR), Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - M Romero
- Cátedra de Física, Facultad de Química, DETEMA, Universidad de la República (UdelaR), General Flores, 2124, 11800, Montevideo, Uruguay
| | - G Grazioli
- Cátedra de Materiales Dentales, Facultad de Odontología, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - H Pérez-Campos
- Instituto Nacional de Donación y Trasplante (INDT), Ministerio de salud Pública-Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Ministerio, Uruguay
| | - A Francia
- Fisiología general y bucodental, Facultad de Odontología, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - W Vicentino
- Instituto Nacional de Donación y Trasplante (INDT), Ministerio de salud Pública-Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Ministerio, Uruguay
| | - A W Mombrú
- Cátedra de Física, Facultad de Química, DETEMA, Universidad de la República (UdelaR), General Flores, 2124, 11800, Montevideo, Uruguay
| | - R Faccio
- Cátedra de Física, Facultad de Química, DETEMA, Universidad de la República (UdelaR), General Flores, 2124, 11800, Montevideo, Uruguay
| | - I Álvarez
- Instituto Nacional de Donación y Trasplante (INDT), Ministerio de salud Pública-Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Ministerio, Uruguay
| | - C Touriño
- Área Terapia Celular y Medicina Regenerativa (ATCMR), Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Uruguay.
| | - H Pardo
- Cátedra de Física, Facultad de Química, DETEMA, Universidad de la República (UdelaR), General Flores, 2124, 11800, Montevideo, Uruguay.
| |
Collapse
|
13
|
Fan X, Sun AR, Young RSE, Afara IO, Hamilton BR, Ong LJY, Crawford R, Prasadam I. Spatial analysis of the osteoarthritis microenvironment: techniques, insights, and applications. Bone Res 2024; 12:7. [PMID: 38311627 PMCID: PMC10838951 DOI: 10.1038/s41413-023-00304-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/21/2023] [Accepted: 11/27/2023] [Indexed: 02/06/2024] Open
Abstract
Osteoarthritis (OA) is a debilitating degenerative disease affecting multiple joint tissues, including cartilage, bone, synovium, and adipose tissues. OA presents diverse clinical phenotypes and distinct molecular endotypes, including inflammatory, metabolic, mechanical, genetic, and synovial variants. Consequently, innovative technologies are needed to support the development of effective diagnostic and precision therapeutic approaches. Traditional analysis of bulk OA tissue extracts has limitations due to technical constraints, causing challenges in the differentiation between various physiological and pathological phenotypes in joint tissues. This issue has led to standardization difficulties and hindered the success of clinical trials. Gaining insights into the spatial variations of the cellular and molecular structures in OA tissues, encompassing DNA, RNA, metabolites, and proteins, as well as their chemical properties, elemental composition, and mechanical attributes, can contribute to a more comprehensive understanding of the disease subtypes. Spatially resolved biology enables biologists to investigate cells within the context of their tissue microenvironment, providing a more holistic view of cellular function. Recent advances in innovative spatial biology techniques now allow intact tissue sections to be examined using various -omics lenses, such as genomics, transcriptomics, proteomics, and metabolomics, with spatial data. This fusion of approaches provides researchers with critical insights into the molecular composition and functions of the cells and tissues at precise spatial coordinates. Furthermore, advanced imaging techniques, including high-resolution microscopy, hyperspectral imaging, and mass spectrometry imaging, enable the visualization and analysis of the spatial distribution of biomolecules, cells, and tissues. Linking these molecular imaging outputs to conventional tissue histology can facilitate a more comprehensive characterization of disease phenotypes. This review summarizes the recent advancements in the molecular imaging modalities and methodologies for in-depth spatial analysis. It explores their applications, challenges, and potential opportunities in the field of OA. Additionally, this review provides a perspective on the potential research directions for these contemporary approaches that can meet the requirements of clinical diagnoses and the establishment of therapeutic targets for OA.
Collapse
Affiliation(s)
- Xiwei Fan
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Antonia Rujia Sun
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Reuben S E Young
- Central Analytical Research Facility, Queensland University of Technology, Brisbane, QLD, Australia
- Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Isaac O Afara
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
- School of Electrical Engineering and Computer Science, Faculty of Engineering, Architecture and Information Technology, University of Queensland, Brisbane, QLD, Australia
| | - Brett R Hamilton
- Centre for Microscopy and Microanalysis, University of Queensland, Brisbane, QLD, Australia
| | - Louis Jun Ye Ong
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Ross Crawford
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
- The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Indira Prasadam
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia.
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia.
| |
Collapse
|
14
|
Batool S, Roth BJ, Xia Y. Depth-Dependent Strain Model (1D) for Anisotropic Fibrils in Articular Cartilage. MATERIALS (BASEL, SWITZERLAND) 2024; 17:238. [PMID: 38204091 PMCID: PMC10779946 DOI: 10.3390/ma17010238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 12/21/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024]
Abstract
The mechanical response of articular cartilage (AC) under compression is anisotropic and depth-dependent. AC is osmotically active, and its intrinsic osmotic swelling pressure is balanced by its collagen fibril network. This mechanism requires the collagen fibers to be under a state of tensile pre-strain. A simple mathematical model is used to explain the depth-dependent strain calculations observed in articular cartilage under 1D axial compression (perpendicular to the articular surface). The collagen fibers are under pre-strain, influenced by proteoglycan concentration (fixed charged density, FCD) and collagen stiffness against swelling stress. The stiffness is introduced in our model as an anisotropic modulus that varies with fibril orientation through tissue depth. The collagen fibers are stiffer to stretching parallel to their length than perpendicular to it; when combined with depth-varying FCD, the model successfully predicts how tissue strains decrease with depth during compression. In summary, this model highlights that the mechanical properties of cartilage depend not only on proteoglycan concentration but also on the intrinsic properties of the pre-strained collagen network. These properties are essential for the proper functioning of articular cartilage.
Collapse
Affiliation(s)
| | - Bradley J. Roth
- Department of Physics, Oakland University, Rochester, MI 48309, USA; (S.B.); (Y.X.)
| | | |
Collapse
|
15
|
Haessler A, Jung N, Windbergs M. Unraveling Molecular Composition in Biological Samples-Systematic Evaluation of Statistical Methods for the Analysis of Hyperspectral Raman Data. Anal Chem 2023; 95:17646-17653. [PMID: 37989265 DOI: 10.1021/acs.analchem.3c03368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
Recently, confocal Raman microscopy has gained popularity in biomedical research for studying tissues in healthy and diseased state due to its ability to acquire chemically selective data in a noninvasive approach. However, biological samples, such as brain tissue, are inherently difficult to analyze due to the superposition of molecules in the Raman spectra and low variation of spectral features within the sample. The analysis is further impeded by pathological hallmarks, for example beta-amyloid (Aβ) plaques in Alzheimer's disease, which are often solely characterized by subtle shifts in the respective Raman peaks. To unravel the underlying molecular information, convoluted statistical procedures are inevitable. Unfortunately, such statistical methods are often inadequately described, and most natural scientists lack knowledge of their appropriate use, causing unreproducible results and stagnation in the application of hyperspectral Raman imaging. Therefore, we have set out to provide a comprehensive guide to address these challenges with the example of a complex hyperspectral data set of brain tissue samples with Aβ plaques. Our study encompasses established as well as novel statistical methods, including univariate analysis, principal component analysis, cluster analysis, spectral unmixing, and 2D correlation spectroscopy, and critically compares the outcomes of each analysis. Moreover, we transparently demonstrate the effect of preprocessing decisions like denoising and scaling techniques, providing valuable insights into implications of spectral quality for data evaluation. Thereby, this study provides a comprehensive evaluation of analysis approaches for complex hyperspectral Raman data, laying out a blueprint for elucidating meaningful information from biological samples in chemical imaging.
Collapse
Affiliation(s)
- Annika Haessler
- Institute of Pharmaceutical Technology, Goethe University Frankfurt am Main, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany
| | - Nathalie Jung
- Institute of Pharmaceutical Technology, Goethe University Frankfurt am Main, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany
| | - Maike Windbergs
- Institute of Pharmaceutical Technology, Goethe University Frankfurt am Main, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany
| |
Collapse
|
16
|
LaLone V, Smith D, Diaz-Espinosa J, Rosania GR. Quantitative Raman chemical imaging of intracellular drug-membrane aggregates and small molecule drug precipitates in cytoplasmic organelles. Adv Drug Deliv Rev 2023; 202:115107. [PMID: 37769851 PMCID: PMC10841539 DOI: 10.1016/j.addr.2023.115107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/02/2023]
Abstract
Raman confocal microscopes have been used to visualize the distribution of small molecule drugs within different subcellular compartments. This visualization allows the discovery, characterization, and detailed analysis of the molecular transport phenomena underpinning the Volume of Distribution - a key parameter governing the systemic pharmacokinetics of small molecule drugs. In the specific case of lipophilic small molecules with large Volumes of Distribution, chemical imaging studies using Raman confocal microscopes have revealed how weakly basic, poorly soluble drug molecules can accumulate inside cells by forming stable, supramolecular complexes in association with cytoplasmic membranes or by precipitating out within organelles. To study the self-assembly and function of the resulting intracellular drug inclusions, Raman chemical imaging methods have been developed to measure and map the mass, concentration, and ionization state of drug molecules at a microscopic, subcellular level. Beyond the field of drug delivery, Raman chemical imaging techniques relevant to the study of microscopic drug precipitates and drug-lipid complexes which form inside cells are also being developed by researchers with seemingly unrelated scientific interests. Highlighting advances in data acquisition, calibration methods, and computational data management and analysis tools, this review will cover a decade of technological developments that enable the conversion of spectral signals obtained from Raman confocal microscopes into new discoveries and information about previously unknown, concentrative drug transport pathways driven by soluble-to-insoluble phase transitions occurring within the cytoplasmic organelles of eukaryotic cells.
Collapse
Affiliation(s)
- Vernon LaLone
- Cambium Analytica Research Laboratories, Traverse City, MI, United States
| | - Doug Smith
- Cambium Analytica Research Laboratories, Traverse City, MI, United States
| | - Jennifer Diaz-Espinosa
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
| | - Gus R Rosania
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
17
|
Shehata E, Nippolainen E, Shaikh R, Ronkainen AP, Töyräs J, Sarin JK, Afara IO. Raman Spectroscopy and Machine Learning Enables Estimation of Articular Cartilage Structural, Compositional, and Functional Properties. Ann Biomed Eng 2023; 51:2301-2312. [PMID: 37328704 PMCID: PMC10518284 DOI: 10.1007/s10439-023-03271-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 06/01/2023] [Indexed: 06/18/2023]
Abstract
OBJECTIVE To differentiate healthy from artificially degraded articular cartilage and estimate its structural, compositional, and functional properties using Raman spectroscopy (RS). DESIGN Visually normal bovine patellae (n = 12) were used in this study. Osteochondral plugs (n = 60) were prepared and artificially degraded either enzymatically (via Collagenase D or Trypsin) or mechanically (via impact loading or surface abrasion) to induce mild to severe cartilage damage; additionally, control plugs were prepared (n = 12). Raman spectra were acquired from the samples before and after artificial degradation. Afterwards, reference biomechanical properties, proteoglycan (PG) content, collagen orientation, and zonal (%) thickness of the samples were measured. Machine learning models (classifiers and regressors) were then developed to discriminate healthy from degraded cartilage based on their Raman spectra and to predict the aforementioned reference properties. RESULTS The classifiers accurately categorized healthy and degraded samples (accuracy = 86%), and successfully discerned moderate from severely degraded samples (accuracy = 90%). On the other hand, the regression models estimated cartilage biomechanical properties with reasonable error (≤ 24%), with the lowest error observed in the prediction of instantaneous modulus (12%). With zonal properties, the lowest prediction errors were observed in the deep zone, i.e., PG content (14%), collagen orientation (29%), and zonal thickness (9%). CONCLUSION RS is capable of discriminating between healthy and damaged cartilage, and can estimate tissue properties with reasonable errors. These findings demonstrate the clinical potential of RS.
Collapse
Affiliation(s)
- Eslam Shehata
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
- Diagnostic Imaging Center, Kuopio University Hospital, Kuopio, Finland
| | - Ervin Nippolainen
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| | - Rubina Shaikh
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| | | | - Juha Töyräs
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
- Science Service Center, Kuopio University Hospital, Kuopio, Finland
- School of Information Technology and Electrical Engineering, The University of Queensland, Brisbane, Australia
| | - Jaakko K. Sarin
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
- Department of Medical Physics, Medical Imaging Center, Pirkanmaa Hospital District, Tampere, Finland
| | - Isaac O. Afara
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
- School of Information Technology and Electrical Engineering, The University of Queensland, Brisbane, Australia
| |
Collapse
|
18
|
Zielinska D, Yosef HK, Zollitsch T, Kern J, Jakob Y, Gvaramia D, Rotter N, Pontiggia L, Moehrlen U, Biedermann T, Klar AS. Characterization of Distinct Chondrogenic Cell Populations of Patients Suffering from Microtia Using Single-Cell Micro-Raman Spectroscopy. Biomedicines 2023; 11:2588. [PMID: 37761029 PMCID: PMC10526501 DOI: 10.3390/biomedicines11092588] [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: 07/06/2023] [Revised: 08/29/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Microtia is a congenital condition of abnormal development of the outer ear. Tissue engineering of the ear is an alternative treatment option for microtia patients. However, for this approach, the identification of high regenerative cartilage progenitor cells is of vital importance. Raman analysis provides a novel, non-invasive, label-free diagnostic tool to detect distinctive biochemical features of single cells or tissues. Using micro-Raman spectroscopy, we were able to distinguish and characterize the particular molecular fingerprints of differentiated chondrocytes and perichondrocytes and their respective progenitors isolated from healthy individuals and microtia patients. We found that microtia chondrocytes exhibited lower lipid concentrations in comparison to healthy cells, thus indicating the importance of fat storage. Moreover, we suggest that collagen is a useful biomarker for distinguishing between populations obtained from the cartilage and perichondrium because of the higher spectral contributions of collagen in the chondrocytes compared to perichondrocytes from healthy individuals and microtia patients. Our results represent a contribution to the identification of cell markers that may allow the selection of specific cell populations for cartilage tissue engineering. Moreover, the observed differences between microtia and healthy cells are essential for gaining better knowledge of the cause of microtia. It can be useful for designing novel treatment options based on further investigations of the discovered biochemical substrate alterations.
Collapse
Affiliation(s)
- Dominika Zielinska
- Tissue Biology Research Unit, University Children’s Hospital Zurich, 8952 Schlieren, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
- Faculty of Medicine, University of Zurich, 8032 Zurich, Switzerland
| | - Hesham K. Yosef
- Faculty of Medicine, University of Zurich, 8032 Zurich, Switzerland
- microphotonXGmbH, 82327 Tutzing, Germany
| | | | - Johann Kern
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Yvonne Jakob
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - David Gvaramia
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Nicole Rotter
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Luca Pontiggia
- Tissue Biology Research Unit, University Children’s Hospital Zurich, 8952 Schlieren, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
- Faculty of Medicine, University of Zurich, 8032 Zurich, Switzerland
| | - Ueli Moehrlen
- Tissue Biology Research Unit, University Children’s Hospital Zurich, 8952 Schlieren, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
- Faculty of Medicine, University of Zurich, 8032 Zurich, Switzerland
- Department of Surgery, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
| | - Thomas Biedermann
- Tissue Biology Research Unit, University Children’s Hospital Zurich, 8952 Schlieren, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
- Faculty of Medicine, University of Zurich, 8032 Zurich, Switzerland
| | - Agnes S. Klar
- Tissue Biology Research Unit, University Children’s Hospital Zurich, 8952 Schlieren, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
- Faculty of Medicine, University of Zurich, 8032 Zurich, Switzerland
| |
Collapse
|
19
|
Raj P, Wu L, Almeida C, Conway L, Tanwar S, Middendorf J, Barman I. Shining Light on Osteoarthritis: Spatially Offset Raman Spectroscopy as a Window into Cartilage Health. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.14.553328. [PMID: 37645996 PMCID: PMC10462085 DOI: 10.1101/2023.08.14.553328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Articular cartilage is a complex tissue, and early detection of osteoarthritis (OA) is crucial for effective treatment. However, current imaging modalities lack molecular specificity and primarily detect late-stage changes. In this study, we propose the use of Spatially Offset Raman Spectroscopy (SORS) for non-invasive, depth-dependent, and molecular-specific diagnostics of articular cartilage. We demonstrate the potential of SORS to penetrate deep layers of cartilage, providing a comprehensive understanding of disease progression. Our SORS measurements were characterized and validated through mechanical and histological techniques, revealing strong correlations between spectroscopic measurements and both Young's modulus and depth of cartilage damage. By longitudinally monitoring enzymatically degraded condyles, we further developed a depth-dependent damage-tracking method. Our analysis revealed distinct components related to sample depth and glycosaminoglycan (GAG) changes, offering a comprehensive picture of cartilage health. Collectively, these findings highlight the potential of SORS as a valuable tool for enhancing OA management and improving patient outcomes.
Collapse
Affiliation(s)
- Piyush Raj
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Lintong Wu
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Craig Almeida
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Lauren Conway
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Swati Tanwar
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jill Middendorf
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ishan Barman
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University, School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
20
|
Zhou H, Llanes JP, Lotfi M, Sarntinoranont M, Simmons CS, Subhash G. Label-Free Quantification of Microscopic Alignment in Engineered Tissue Scaffolds by Polarized Raman Spectroscopy. ACS Biomater Sci Eng 2023; 9:3206-3218. [PMID: 37170804 DOI: 10.1021/acsbiomaterials.3c00242] [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] [Indexed: 05/13/2023]
Abstract
Monitoring of extracellular matrix (ECM) microstructure is essential in studying structure-associated cellular processes, improving cellular function, and for ensuring sufficient mechanical integrity in engineered tissues. This paper describes a novel method to study the microscale alignment of the matrix in engineered tissue scaffolds (ETS) that are usually composed of a variety of biomacromolecules derived by cells. First, a trained loading function was derived from Raman spectra of highly aligned native tissue via principal component analysis (PCA), where prominent changes associated with specific Raman bands (e.g., 1444, 1465, 1605, 1627-1660, and 1665-1689 cm-1) were detected with respect to the polarization angle. These changes were mainly caused by the aligned matrix of many compounds within the tissue relative to the laser polarization, including proteins, lipids, and carbohydrates. Hence this trained function was applied to quantify the alignment within ETS of various matrix components derived by cells. Furthermore, a simple metric called Amplitude Alignment Metric (AAM) was derived to correlate the orientation dependence of polarized Raman spectra of ETS to the degree of matrix alignment. It was found that the AAM was significantly higher in anisotropic ETS than isotropic ones. The PRS method revealed a lower p-value for distinguishing the alignment between these two types of ETS as compared to the microscopic method for detecting fluorescent-labeled protein matrices at a similar microscopic scale. These results indicate that the anisotropy of a complex matrix in engineered tissue can be assessed at the microscopic scale using a PRS-based simple metric, which is superior to the traditional microscopic method. This PRS-based method can serve as a complementary tool for the design and assessment of engineered tissues that mimic the native matrix organizational microstructures.
Collapse
Affiliation(s)
- Hui Zhou
- Mechanical and Aerospace Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Janny Piñeiro Llanes
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Maedeh Lotfi
- Mechanical and Aerospace Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Malisa Sarntinoranont
- Mechanical and Aerospace Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Chelsey S Simmons
- Mechanical and Aerospace Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Ghatu Subhash
- Mechanical and Aerospace Engineering, University of Florida, Gainesville, Florida 32611, United States
| |
Collapse
|
21
|
Liu J, Huang B, Ma Z, Xu S, Zhao H, Ren L. Full Regional Creep Displacement Map of Articular Cartilage Based on Nanoindentation Array. ACS Biomater Sci Eng 2023. [PMID: 37115745 DOI: 10.1021/acsbiomaterials.2c01395] [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: 04/29/2023]
Abstract
The elucidation of the mechanisms underlying articular cartilage lesions poses a formidable challenge in the field of cartilage repair. Despite significant strides in cartilage mechanics research, the region-dependent creep properties of articular cartilage remain elusive. In this study, we employ depth-sensing indentation tests to experimentally determine the creep properties of four distinct regions of articular cartilage, thereby unveiling a region-dependent full map of creep parameters. The measured creep displacement-time response curves indicate that the creep properties of the articular cartilage exhibit a clear regional correlation. Accordingly, the full regional creep map of articular cartilage is experimentally constructed for the first time. The correlation between the microstructures and the creep properties of cartilage in different regions is revealed. A three-parameter model is established to describe the creep velocity-displacement response of cartilage. Raman spectra reveal that the proteoglycan content is positively correlated with creep properties. The Raman shift directly indicates diverse residual stresses in different microregions. The obtained data facilitate a deep understanding of the potential creep dependent damage mechanism of cartilage and the further development of artificial cartilage materials.
Collapse
Affiliation(s)
- Jize Liu
- School of Mechanical and Aerospace Engineering, Jilin University, Changchun 130025, China
| | - Bin Huang
- School of Mechanical and Aerospace Engineering, Jilin University, Changchun 130025, China
| | - Zhichao Ma
- School of Mechanical and Aerospace Engineering, Jilin University, Changchun 130025, China
- Key Laboratory of CNC Equipment Reliability Ministry of Education, Jilin University, Changchun 130025, China
| | - Shuting Xu
- School of Mechanical and Aerospace Engineering, Jilin University, Changchun 130025, China
| | - Hongwei Zhao
- School of Mechanical and Aerospace Engineering, Jilin University, Changchun 130025, China
- Key Laboratory of CNC Equipment Reliability Ministry of Education, Jilin University, Changchun 130025, China
| | - Luquan Ren
- Weihai Institute for Bionics-Jilin University, Weihai 264207, China
| |
Collapse
|
22
|
LaLone V, Aizenshtadt A, Goertz J, Skottvoll FS, Mota MB, You J, Zhao X, Berg HE, Stokowiec J, Yu M, Schwendeman A, Scholz H, Wilson SR, Krauss S, Stevens MM. Quantitative chemometric phenotyping of three-dimensional liver organoids by Raman spectral imaging. CELL REPORTS METHODS 2023; 3:100440. [PMID: 37159662 PMCID: PMC10162950 DOI: 10.1016/j.crmeth.2023.100440] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 02/06/2023] [Accepted: 03/08/2023] [Indexed: 05/11/2023]
Abstract
Confocal Raman spectral imaging (RSI) enables high-content, label-free visualization of a wide range of molecules in biological specimens without sample preparation. However, reliable quantification of the deconvoluted spectra is needed. Here we develop an integrated bioanalytical methodology, qRamanomics, to qualify RSI as a tissue phantom calibrated tool for quantitative spatial chemotyping of major classes of biomolecules. Next, we apply qRamanomics to fixed 3D liver organoids generated from stem-cell-derived or primary hepatocytes to assess specimen variation and maturity. We then demonstrate the utility of qRamanomics for identifying biomolecular response signatures from a panel of liver-altering drugs, probing drug-induced compositional changes in 3D organoids followed by in situ monitoring of drug metabolism and accumulation. Quantitative chemometric phenotyping constitutes an important step in developing quantitative label-free interrogation of 3D biological specimens.
Collapse
Affiliation(s)
- Vernon LaLone
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, UK
- Hybrid Technology Hub-Centre of Excellence, Imperial College London, London SW7 2AZ, UK
| | - Aleksandra Aizenshtadt
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, P.O. Box 1112, Blindern, 0317 Oslo, Norway
| | - John Goertz
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, UK
| | - Frøydis Sved Skottvoll
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, P.O. Box 1112, Blindern, 0317 Oslo, Norway
- Department of Chemistry, University of Oslo, P.O. Box 1033, Blindern, 0315 Oslo, Norway
| | - Marco Barbero Mota
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, UK
| | - Junji You
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, UK
| | - Xiaoyu Zhao
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, UK
| | - Henriette Engen Berg
- Department of Chemistry, University of Oslo, P.O. Box 1033, Blindern, 0315 Oslo, Norway
| | - Justyna Stokowiec
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, P.O. Box 1112, Blindern, 0317 Oslo, Norway
| | - Minzhi Yu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hanne Scholz
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, P.O. Box 1112, Blindern, 0317 Oslo, Norway
- Department of Transplant Medicine, Oslo University Hospital, Oslo, Norway
- Institute for Surgical Research, Oslo University Hospital, Oslo, Norway
| | - Steven Ray Wilson
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, P.O. Box 1112, Blindern, 0317 Oslo, Norway
- Department of Chemistry, University of Oslo, P.O. Box 1033, Blindern, 0315 Oslo, Norway
| | - Stefan Krauss
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, P.O. Box 1112, Blindern, 0317 Oslo, Norway
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, P.O. Box 4950, Nydalen, 0424 Oslo, Norway
| | - Molly M. Stevens
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, UK
- Hybrid Technology Hub-Centre of Excellence, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
23
|
Zhang S, Chen S, Zhu R. Electroporation-Assisted Surface-Enhanced Raman Detection for Long-Term, Label-Free, and Noninvasive Molecular Profiling of Live Single Cells. ACS Sens 2023; 8:555-564. [PMID: 36399395 DOI: 10.1021/acssensors.2c01582] [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: 11/19/2022]
Abstract
Molecule characterization of live single cells is greatly important in disease diagnoses and personalized treatments. Conventional molecule detection methods, such as mass spectrography, gene sequencing, or immunofluorescence, are usually destructive or labeled and unable to monitor the dynamic change of live cellular molecules. Herein, we propose an electroporation-assisted surface-enhanced Raman scattering (EP-SERS) method using a microchip to implement label-free, noninvasive, and continuous detections of the molecules of live single cells. The microchip containing microelectrodes with nanostructured EP-SERS probes has a multifunction of cell positioning, electroporation, and SERS detection. The EP-SERS method capably detects both the intracellular and extracellular molecules of live single cells without losing cell viability so as to enable long-term monitoring of the molecular pathological process in situ. We detect the molecules of single cells for two breast cancer cell lines with different malignancies (MCF-7 and MDA-MB-231), one liver cancer cell line (Huh-7), and one normal cell line (293T) using the EP-SERS method and classify these cell types to achieve high accuracies of 91.4-98.3% using their SERS spectra. Furthermore, 24 h continuous monitoring of the heterogeneous molecular responses of different cancer cell lines under doxorubicin treatment is successfully implemented using the EP-SERS method. This work provides a long-term, label-free, and biocompatible approach to simultaneously detect intracellular and extracellular molecules of live single cells on a chip, which would facilitate research and applications of cancer diagnoses and personalized treatments.
Collapse
Affiliation(s)
- Shengsen Zhang
- State Key Laboratory of Precision Measurement Technology and Instrument, Department of Precision Instrument, Tsinghua University, Beijing100084, China
| | - Shengjie Chen
- State Key Laboratory of Precision Measurement Technology and Instrument, Department of Precision Instrument, Tsinghua University, Beijing100084, China
| | - Rong Zhu
- State Key Laboratory of Precision Measurement Technology and Instrument, Department of Precision Instrument, Tsinghua University, Beijing100084, China
| |
Collapse
|
24
|
Anthis AHC, Abundo MP, Neuer AL, Tsolaki E, Rosendorf J, Rduch T, Starsich FHL, Weisse B, Liska V, Schlegel AA, Shapiro MG, Herrmann IK. Modular stimuli-responsive hydrogel sealants for early gastrointestinal leak detection and containment. Nat Commun 2022; 13:7311. [PMID: 36437258 PMCID: PMC9701692 DOI: 10.1038/s41467-022-34272-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 10/19/2022] [Indexed: 11/28/2022] Open
Abstract
Millions of patients every year undergo gastrointestinal surgery. While often lifesaving, sutured and stapled reconnections leak in around 10% of cases. Currently, surgeons rely on the monitoring of surrogate markers and clinical symptoms, which often lack sensitivity and specificity, hence only offering late-stage detection of fully developed leaks. Here, we present a holistic solution in the form of a modular, intelligent suture support sealant patch capable of containing and detecting leaks early. The pH and/or enzyme-responsive triggerable sensing elements can be read out by point-of-need ultrasound imaging. We demonstrate reliable detection of the breaching of sutures, in as little as 3 hours in intestinal leak scenarios and 15 minutes in gastric leak conditions. This technology paves the way for next-generation suture support materials that seal and offer disambiguation in cases of anastomotic leaks based on point-of-need monitoring, without reliance on complex electronics or bulky (bio)electronic implantables.
Collapse
Affiliation(s)
- Alexandre H C Anthis
- Nanoparticle Systems Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Sonneggstrasse 3, CH-8092, Zurich, Switzerland
- Laboratory for Particles Biology Interactions, Department Materials Meet Life, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5, CH-9014, St. Gallen, Switzerland
| | - Maria Paulene Abundo
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Anna L Neuer
- Nanoparticle Systems Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Sonneggstrasse 3, CH-8092, Zurich, Switzerland
- Laboratory for Particles Biology Interactions, Department Materials Meet Life, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5, CH-9014, St. Gallen, Switzerland
| | - Elena Tsolaki
- Nanoparticle Systems Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Sonneggstrasse 3, CH-8092, Zurich, Switzerland
- Laboratory for Particles Biology Interactions, Department Materials Meet Life, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5, CH-9014, St. Gallen, Switzerland
| | - Jachym Rosendorf
- Department of Surgery, Faculty of Medicine in Pilsen, Charles University, Prague, Czech Republic
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Prague, Czech Republic
| | - Thomas Rduch
- Laboratory for Particles Biology Interactions, Department Materials Meet Life, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5, CH-9014, St. Gallen, Switzerland
- Department of Gynaecology, Cantonal Hospital St Gallen (KSSG), Rorschacherstrasse 95, CH-9007, St Gallen, Switzerland
| | - Fabian H L Starsich
- Nanoparticle Systems Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Sonneggstrasse 3, CH-8092, Zurich, Switzerland
- Laboratory for Particles Biology Interactions, Department Materials Meet Life, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5, CH-9014, St. Gallen, Switzerland
| | - Bernhard Weisse
- Laboratory for Mechanical Systems Engineering, Department of Engineering Sciences, Empa - Swiss Laboratories for Materials Science and Technology, Ueberlandstrasse 129, CH-8600, Dübendorf, Switzerland
| | - Vaclav Liska
- Department of Surgery, Faculty of Medicine in Pilsen, Charles University, Prague, Czech Republic
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Prague, Czech Republic
| | - Andrea A Schlegel
- Department of Visceral Surgery and Transplantation, University Hospital Zurich, CH-8091, Zurich, Switzerland
- Swiss HPB and Transplant Center, Zurich, Rämistrasse 100, CH-8091, Zurich, Switzerland
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Centre of Preclinical Research, Milan, 20122, Italy
| | - Mikhail G Shapiro
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
- Howard Hughes Medical Institute, Pasadena, CA, 91125, USA
| | - Inge K Herrmann
- Nanoparticle Systems Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Sonneggstrasse 3, CH-8092, Zurich, Switzerland.
- Laboratory for Particles Biology Interactions, Department Materials Meet Life, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5, CH-9014, St. Gallen, Switzerland.
| |
Collapse
|
25
|
Layered mechanical and electrical properties of porcine articular cartilage. Med Biol Eng Comput 2022; 60:3019-3028. [DOI: 10.1007/s11517-022-02653-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 07/28/2022] [Indexed: 10/14/2022]
|
26
|
Tiffany AS, Harley BAC. Growing Pains: The Need for Engineered Platforms to Study Growth Plate Biology. Adv Healthc Mater 2022; 11:e2200471. [PMID: 35905390 PMCID: PMC9547842 DOI: 10.1002/adhm.202200471] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 07/11/2022] [Indexed: 01/27/2023]
Abstract
Growth plates, or physis, are highly specialized cartilage tissues responsible for longitudinal bone growth in children and adolescents. Chondrocytes that reside in growth plates are organized into three distinct zones essential for proper function. Modeling key features of growth plates may provide an avenue to develop advanced tissue engineering strategies and perspectives for cartilage and bone regenerative medicine applications and a platform to study processes linked to disease progression. In this review, a brief introduction of the growth plates and their role in skeletal development is first provided. Injuries and diseases of the growth plates as well as physiological and pathological mechanisms associated with remodeling and disease progression are discussed. Growth plate biology, namely, its architecture and extracellular matrix organization, resident cell types, and growth factor signaling are then focused. Next, opportunities and challenges for developing 3D biomaterial models to study aspects of growth plate biology and disease in vitro are discussed. Finally, opportunities for increasingly sophisticated in vitro biomaterial models of the growth plate to study spatiotemporal aspects of growth plate remodeling, to investigate multicellular signaling underlying growth plate biology, and to develop platforms that address key roadblocks to in vivo musculoskeletal tissue engineering applications are described.
Collapse
Affiliation(s)
- Aleczandria S. Tiffany
- Department of Chemical and Biomolecular EngineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - Brendan A. C. Harley
- Department of Chemical and Biomolecular EngineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
- Carl R. Woese Institute for Genomic BiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| |
Collapse
|
27
|
Lucas IT, Bazin D, Daudon M. Raman opportunities in the field of pathological calcifications. CR CHIM 2022. [DOI: 10.5802/crchim.110] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
28
|
Walther AR, Ditzel N, Kassem M, Andersen MØ, Hedegaard MAB. In vivo non-invasive monitoring of tissue development in 3D printed subcutaneous bone scaffolds using fibre-optic Raman spectroscopy. BIOMATERIALS AND BIOSYSTEMS 2022; 7:100059. [PMID: 36824488 PMCID: PMC9934492 DOI: 10.1016/j.bbiosy.2022.100059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/04/2022] [Accepted: 07/25/2022] [Indexed: 10/16/2022] Open
Abstract
The development of novel biomaterials for regenerative therapy relies on the ability to assess tissue development, quality, and similarity with native tissue types in in vivo experiments. Non-invasive imaging modalities such as X-ray computed tomography offer high spatial resolution but limited biochemical information while histology and biochemical assays are destructive. Raman spectroscopy is a non-invasive, label-free and non-destructive technique widely applied for biochemical characterization. Here we demonstrate the use of fibre-optic Raman spectroscopy for in vivo quantitative monitoring of tissue development in subcutaneous calcium phosphate scaffolds in mice over 16 weeks. Raman spectroscopy was able to quantify the time dependency of different tissue components related to the presence, absence, and quantity of mesenchymal stem cells. Scaffolds seeded with stem cells produced 3-5 times higher amount of collagen-rich extracellular matrix after 16 weeks implantation compared to scaffolds without. These however, showed a 2.5 times higher amount of lipid-rich tissue compared to implants with stem cells. Ex vivo micro-computed tomography and histology showed stem cell mediated collagen and bone development. Histological measures of collagen correlated well with Raman derived quantifications (correlation coefficient in vivo 0.74, ex vivo 0.93). In the absence of stem cells, the scaffolds were largely occupied by adipocytes. The technique developed here could potentially be adapted for a range of small animal experiments for assessing tissue engineering strategies at the biochemical level.
Collapse
Affiliation(s)
- Anders Runge Walther
- SDU Biotechnology, Department of Green Technology, University of Southern Denmark, Campusvej 55, DK-5230 Odense, Denmark
| | - Nicholas Ditzel
- Endocrine Research (KMEB), Department of Endocrinology, Odense University Hospital and University of Southern Denmark, J.B. Winsløws Vej 25, DK-5000 Odense, Denmark
| | - Moustapha Kassem
- Endocrine Research (KMEB), Department of Endocrinology, Odense University Hospital and University of Southern Denmark, J.B. Winsløws Vej 25, DK-5000 Odense, Denmark
| | - Morten Østergaard Andersen
- SDU Biotechnology, Department of Green Technology, University of Southern Denmark, Campusvej 55, DK-5230 Odense, Denmark
| | - Martin Aage Barsøe Hedegaard
- SDU Biotechnology, Department of Green Technology, University of Southern Denmark, Campusvej 55, DK-5230 Odense, Denmark
| |
Collapse
|
29
|
Adachi T, Miyamoto N, Imamura H, Yamamoto T, Marin E, Zhu W, Kobara M, Sowa Y, Tahara Y, Kanamura N, Akiyoshi K, Mazda O, Nishimura I, Pezzotti G. Three-Dimensional Culture of Cartilage Tissue on Nanogel-Cross-Linked Porous Freeze-Dried Gel Scaffold for Regenerative Cartilage Therapy: A Vibrational Spectroscopy Evaluation. Int J Mol Sci 2022; 23:ijms23158099. [PMID: 35897669 PMCID: PMC9332688 DOI: 10.3390/ijms23158099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 02/01/2023] Open
Abstract
This study presents a set of vibrational characterizations on a nanogel-cross-linked porous freeze-dried gel (NanoCliP-FD gel) scaffold for tissue engineering and regenerative therapy. This scaffold is designed for the in vitro culture of high-quality cartilage tissue to be then transplanted in vivo to enable recovery from congenital malformations in the maxillofacial area or crippling jaw disease. The three-dimensional scaffold for in-plate culture is designed with interface chemistry capable of stimulating cartilage formation and maintaining its structure through counteracting the dedifferentiation of mesenchymal stem cells (MSCs) during the formation of cartilage tissue. The developed interface chemistry enabled high efficiency in both growth rate and tissue quality, thus satisfying the requirements of large volumes, high matrix quality, and superior mechanical properties needed in cartilage transplants. We characterized the cartilage tissue in vitro grown on a NanoCliP-FD gel scaffold by human periodontal ligament-derived stem cells (a type of MSC) with cartilage grown by the same cells and under the same conditions on a conventional (porous) atelocollagen scaffold. The cartilage tissues produced by the MSCs on different scaffolds were comparatively evaluated by immunohistochemical and spectroscopic analyses. Cartilage differentiation occurred at a higher rate when MSCs were cultured on the NanoCliP-FD gel scaffold compared to the atelocollagen scaffold, and produced a tissue richer in cartilage matrix. In situ spectroscopic analyses revealed the cell/scaffold interactive mechanisms by which the NanoCliP-FD gel scaffold stimulated such increased efficiency in cartilage matrix formation. In addition to demonstrating the high potential of human periodontal ligament-derived stem cell cultures on NanoCliP-FD gel scaffolds in regenerative cartilage therapy, the present study also highlights the novelty of Raman spectroscopy as a non-destructive method for the concurrent evaluation of matrix quality and cell metabolic response. In situ Raman analyses on living cells unveiled for the first time the underlying physiological mechanisms behind such improved chondrocyte performance.
Collapse
Affiliation(s)
- Tetsuya Adachi
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan; (N.M.); (H.I.); (T.Y.); (E.M.); (N.K.)
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan; (Y.S.); (O.M.)
- Correspondence: (T.A.); (G.P.)
| | - Nao Miyamoto
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan; (N.M.); (H.I.); (T.Y.); (E.M.); (N.K.)
- Infectious Diseases, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Hayata Imamura
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan; (N.M.); (H.I.); (T.Y.); (E.M.); (N.K.)
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan;
| | - Toshiro Yamamoto
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan; (N.M.); (H.I.); (T.Y.); (E.M.); (N.K.)
| | - Elia Marin
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan; (N.M.); (H.I.); (T.Y.); (E.M.); (N.K.)
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan;
| | - Wenliang Zhu
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan;
| | - Miyuki Kobara
- Department of Clinical Pharmacology, Division of Pathological Science, Kyoto Pharmaceutical University, Misasagi Nakauchi-cho, Yamashina-ku, Kyoto 607-8414, Japan;
| | - Yoshihiro Sowa
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan; (Y.S.); (O.M.)
- Department of Plastic and Reconstructive Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yoshiro Tahara
- Department of Chemical Engineering and Materials Science, Doshisha University, 1-3 Tatara Miyakodani, Kyotanabe, Kyoto 610-0394, Japan;
| | - Narisato Kanamura
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan; (N.M.); (H.I.); (T.Y.); (E.M.); (N.K.)
| | - Kazunari Akiyoshi
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan;
| | - Osam Mazda
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan; (Y.S.); (O.M.)
| | - Ichiro Nishimura
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA 90095, USA;
- Division of Advanced Prosthodontics, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA 90095, USA
| | - Giuseppe Pezzotti
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan; (N.M.); (H.I.); (T.Y.); (E.M.); (N.K.)
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan; (Y.S.); (O.M.)
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan;
- Biomedical Research Center, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan
- Correspondence: (T.A.); (G.P.)
| |
Collapse
|
30
|
Cui A, Nippolainen E, Shaikh R, Torniainen J, Ristaniemi A, Finnilä M, Korhonen RK, Saarakkala S, Herzog W, Töyräs J, Afara IO. Assessment of Ligament Viscoelastic Properties Using Raman Spectroscopy. Ann Biomed Eng 2022; 50:1134-1142. [PMID: 35802206 PMCID: PMC9363474 DOI: 10.1007/s10439-022-02988-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 05/31/2022] [Indexed: 11/01/2022]
Abstract
Injuries to the ligaments of the knee commonly impact vulnerable and physically active individuals. These injuries can lead to the development of degenerative diseases such as post-traumatic osteoarthritis (PTOA). Non-invasive optical modalities, such as infrared and Raman spectroscopy, provide means for quantitative evaluation of knee joint tissues and have been proposed as potential quantitative diagnostic tools for arthroscopy. In this study, we evaluate Raman spectroscopy as a viable tool for estimating functional properties of collateral ligaments. Artificial trauma was induced by anterior cruciate ligament transection (ACLT) in the left or right knee joint of skeletally mature New Zealand rabbits. The corresponding contralateral (CL) samples were extracted from healthy unoperated joints along with a separate group of control (CNTRL) animals. The rabbits were sacrificed at 8 weeks after ACLT. The ligaments were then harvested and measured using Raman spectroscopy. A uniaxial tensile stress-relaxation testing protocol was adopted for determining several biomechanical properties of the samples. Partial least squares (PLS) regression models were then employed to correlate the spectral data with the biomechanical properties. Results show that the capacity of Raman spectroscopy for estimating the biomechanical properties of the ligament samples varies depending on the target property, with prediction error ranging from 15.78% for tissue cross-sectional area to 30.39% for stiffness. The hysteresis under cyclic loading at 2 Hz (RMSE = 6.22%, Normalized RMSE = 22.24%) can be accurately estimated from the Raman data which describes the viscous damping properties of the tissue. We conclude that Raman spectroscopy has the potential for non-destructively estimating ligament biomechanical properties in health and disease, thus enhancing the diagnostic value of optical arthroscopic evaluations of ligament integrity.
Collapse
Affiliation(s)
- Andy Cui
- School of Information Technology and Electrical Engineering, The University of Queensland, Brisbane, Australia.
| | - Ervin Nippolainen
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| | - Rubina Shaikh
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland.,Department of Orthopedics, Traumatology and Hand Surgery, Kuopio University Hospital, Kuopio, Finland
| | - Jari Torniainen
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| | - Aapo Ristaniemi
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland.,AO Research Institute Davos, Davos, Switzerland
| | - Mikko Finnilä
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Rami K Korhonen
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| | - Simo Saarakkala
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland.,Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland
| | - Walter Herzog
- Human Performance Lab, Faculty of Kinesiology, University of Calgary, Calgary, Canada
| | - Juha Töyräs
- School of Information Technology and Electrical Engineering, The University of Queensland, Brisbane, Australia.,Department of Applied Physics, University of Eastern Finland, Kuopio, Finland.,Science Service Center, Kuopio University Hospital, Kuopio, Finland
| | - Isaac O Afara
- School of Information Technology and Electrical Engineering, The University of Queensland, Brisbane, Australia.,Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
31
|
Jaabar IL, Cornette P, Miche A, Wanherdrick K, Dupres V, Ehkirch FP, Cambon Binder A, Berenbaum F, Houard X, Landoulsi J. Deciphering pathological remodelling of the human cartilage extracellular matrix in osteoarthritis at the supramolecular level. NANOSCALE 2022; 14:8691-8708. [PMID: 35673929 DOI: 10.1039/d2nr00474g] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The extracellular matrix (ECM) of articular cartilage is a three-dimensional network mainly constituted of entangled collagen fibrils and interfibrillar aggrecan aggregates. During the development of osteoarthritis (OA), the most common musculoskeletal disorder, the ECM is subjected to a combination of chemical and structural changes that play a pivotal role in the initiation and the progress of the disease. While the molecular mechanisms involved in the pathological remodelling of the ECM are considered as decisive, they remain, however, not completely elucidated. Herein, we report a relevant way for unravelling the role and nature of OA progress on human cartilage tissues, in terms of chemical composition and morphological and mechanical properties at the level of supramolecular assemblies constituting the cartilage ECM. For this purpose, we used X-ray photoelectron spectroscopy (XPS), and developed an innovative methodological approach that provides the molecular composition of the ECM. Moreover, we used atomic force microscopy (AFM) to probe the tissues at the level of individual collagen fibrils, both imaging and force spectroscopy modes being explored to this end. Taken together, these nanoscale characterization studies reveal the existence of two stages in the OA progress. At the early stage, a marked increase in the aggrecan and collagen content is observed, reflecting the homeostatic chondrocyte activity that tends to repair the cartilage ECM. At the late stage, we observe a failed attempt to stabilize and/or restore the tissue, yielding significant degradation of the supramolecular assemblies. This suggests an imbalance in the chondrocyte activity that turns in favor of catabolic events. Chemical changes are also accompanied by ECM structural changes and stiffening. Interestingly, we showed the possibility to mimic the imbalanced activities of chondrocytes by applying enzymatic digestions of healthy cartilage, through the combined action of hyaluronidase and collagenase. This yields damage strictly analogous to that observed at high OA severity. These findings bring mechanistic insights leading to a better understanding of the mechanism by which OA is initiated and progresses in the cartilage ECM. They offer guidelines for the development of curative treatments, such as targeting the homeostatic balance of chondrocyte metabolism through the control of enzymatic reactions involved in catabolic processes.
Collapse
Affiliation(s)
- Ilhem Lilia Jaabar
- Sorbonne Université, CNRS, Laboratoire de Réactivité de Surface, LRS, F-75005 Paris, France.
- Sorbonne Université, INSERM (UMR_S938), Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.
| | - Pauline Cornette
- Sorbonne Université, CNRS, Laboratoire de Réactivité de Surface, LRS, F-75005 Paris, France.
- Sorbonne Université, INSERM (UMR_S938), Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.
| | - Antoine Miche
- Sorbonne Université, CNRS, Laboratoire de Réactivité de Surface, LRS, F-75005 Paris, France.
| | - Kristell Wanherdrick
- Sorbonne Université, INSERM (UMR_S938), Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.
| | - Vincent Dupres
- Université Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000, Lille, France
| | - François-Paul Ehkirch
- Sorbonne Université, INSERM (UMR_S938), Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.
- Clinique Maussins-Nollet, F-75019 Paris, France
| | - Adeline Cambon Binder
- Sorbonne Université, INSERM (UMR_S938), Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.
- Orthopedics and Hand Surgery Department, Saint-Antoine Hospital, 184 Rue du Faubourg Saint-Antoine, Paris, 75012, France
| | - Francis Berenbaum
- Sorbonne Université, INSERM (UMR_S938), Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.
- Rheumatology Department, AP-HP Saint-Antoine Hospital, 184, rue du Faubourg Saint-Antoine, 75012, Paris, France
| | - Xavier Houard
- Sorbonne Université, INSERM (UMR_S938), Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.
| | - Jessem Landoulsi
- Sorbonne Université, CNRS, Laboratoire de Réactivité de Surface, LRS, F-75005 Paris, France.
| |
Collapse
|
32
|
Zheng W, Xie R, Liang X, Liang Q. Fabrication of Biomaterials and Biostructures Based On Microfluidic Manipulation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2105867. [PMID: 35072338 DOI: 10.1002/smll.202105867] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 12/22/2021] [Indexed: 06/14/2023]
Abstract
Biofabrication technologies are of importance for the construction of organ models and functional tissue replacements. Microfluidic manipulation, a promising biofabrication technique with micro-scale resolution, can not only help to realize the fabrication of specific microsized structures but also build biomimetic microenvironments for biofabricated tissues. Therefore, microfluidic manipulation has attracted attention from researchers in the manipulation of particles and cells, biochemical analysis, tissue engineering, disease diagnostics, and drug discovery. Herein, biofabrication based on microfluidic manipulation technology is reviewed. The application of microfluidic manipulation technology in the manufacturing of biomaterials and biostructures with different dimensions and the control of the microenvironment is summarized. Finally, current challenges are discussed and a prospect of microfluidic manipulation technology is given. The authors hope this review can provide an overview of microfluidic manipulation technologies used in biofabrication and thus steer the current efforts in this field.
Collapse
Affiliation(s)
- Wenchen Zheng
- Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Ruoxiao Xie
- Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Xiaoping Liang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangdong, 510006, China
| | - Qionglin Liang
- Department of Chemistry, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
33
|
Terpstra ML, Li J, Mensinga A, de Ruijter M, van Rijen MHP, Androulidakis C, Galiotis C, Papantoniou I, Matsusaki M, Malda J, Levato R. Bioink with cartilage-derived extracellular matrix microfibers enables spatial control of vascular capillary formation in bioprinted constructs. Biofabrication 2022; 14. [PMID: 35354130 DOI: 10.1088/1758-5090/ac6282] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 03/30/2022] [Indexed: 11/11/2022]
Abstract
Microvasculature is essential for the exchange of gas and nutrient for most tissues in our body. Some tissue structures such as the meniscus presents spatially confined blood vessels adjacent to non-vascularized regions. In biofabrication, mimicking the spatial distribution of such vascular components is paramount, as capillary ingrowth into non-vascularized tissues can lead to tissue matrix alterations and subsequent pathology. Multi-material 3D bioprinting can potentially resolve anisotropic tissue features, although building complex constructs comprising stable vascularized and non-vascularized regions remains a major challenge. Here, we developed endothelial cell(EC)-laden pro- and anti-angiogenic bioinks, supplemented with bioactive matrix-derived microfibers (MFs) that were created from type I collagen sponges (col-1) and cartilage decellularized extracellular matrix (CdECM). EC-driven capillary network formation started two days after bioprinting. Supplementing cartilage-derived MFs to endothelial-cell laden bioinks reduced the total length of neo-microvessels by 29% after 14 days, compared to col-1 MFs-laden bioinks. As a proof of concept, the bioinks were bioprinted into an anatomical meniscus shape with a biomimetic vascularized outer and non-vascularized inner region, using a microgel suspension bath. The constructs were cultured up to 14 days, with in the outer zone the HUVEC-, mural cell-, and col-1 MF-laden pro-angiogenic bioink, and in the inner zone a meniscus progenitor cell (MPC)- and CdECM MF-laden anti-angiogenic bioink, revealing successful spatial confinement of the nascent vascular network only in the outer zone. Further, to co-facilitate both microvessel formation and MPC-derived matrix formation, we formulated cell culture medium conditions with a temporal switch. Overall, this study provides a new strategy that could be applied to develop zonal biomimetic meniscal constructs. Moreover, the use of ECM-derived MFs to promote or inhibit capillary networks opens new possibilities for the biofabrication of tissues with anisotropic microvascular distribution. These have potential for many applications including in vitro models, cancer progression, and testing anti-angiogenic therapies.
Collapse
Affiliation(s)
- Margo Luchiena Terpstra
- University Medical Centre Utrecht Department of Orthopedics, Heidelberglaan 100, Utrecht, 3584 CX, NETHERLANDS
| | - Jinyu Li
- Department of Applied Chemistry, Osaka University, Faculty of Engineering, Suita, Osaka 565-0871, Suita, Osaka, 565-0871, JAPAN
| | - Anneloes Mensinga
- Utrecht University Faculty of Veterinary Medicine, Heidelberglaan 8, Utrecht, Utrecht, 3584 CS, NETHERLANDS
| | - Mylène de Ruijter
- University Medical Centre Utrecht Department of Orthopedics, Heidelberglaan 100, Utrecht, Utrecht, 3584 CX, NETHERLANDS
| | - Mattie H P van Rijen
- Department of Orthopedics, Universitair Medisch Centrum Utrecht, Heidelberglaan 100, Utrecht, Utrecht, 3584 CX, NETHERLANDS
| | - Charalampos Androulidakis
- Department of Chemical Engineering, University of Patras, Stadiou Street, Platani, Patras, Periféria Dhitikís Elládh, 26504, GREECE
| | - Costas Galiotis
- Department Chemical EngineeringScience, University of Patras, Panepistimioupoli, Rio, GR-26504 Patras, Patra, Periféria Dhitikís Elládh, 26504, GREECE
| | - Ioannis Papantoniou
- Prometheus Division of Skeletal Tissue Engineering, KU Leuven, Onderwijs en Navorsing 1, +10, Herestraat 49, box 813, Leuven, 3000, BELGIUM
| | - Michiya Matsusaki
- Department of Applied Chemistry, Osaka University, Faculty of Engineering, Suita, Osaka 565-0871, Suita, Osaka, 565-0871, JAPAN
| | - Jos Malda
- Orthopaedics, University Medical Centre Utrecht Department of Orthopedics, The Netherlands, Utrecht, 3508 GA, NETHERLANDS
| | - Riccardo Levato
- Utrecht University Faculty of Veterinary Medicine, Heidelberglaan 8, Utrecht, 3584 CS, NETHERLANDS
| |
Collapse
|
34
|
Elídóttir KL, Scott L, Lewis R, Jurewicz I. Biomimetic approach to articular cartilage tissue engineering using carbon nanotube-coated and textured polydimethylsiloxane scaffolds. Ann N Y Acad Sci 2022; 1513:48-64. [PMID: 35288951 PMCID: PMC9545810 DOI: 10.1111/nyas.14769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 02/18/2022] [Indexed: 11/27/2022]
Abstract
There is a significant need to understand the complexity and heterogeneity of articular cartilage to develop more effective therapeutic strategies for diseases such as osteoarthritis. Here, we show that carbon nanotubes (CNTs) are excellent candidates as a material for synthetic scaffolds to support the growth of chondrocytes—the cells that produce and maintain cartilage. Chondrocyte morphology, proliferation, and alignment were investigated as nanoscale CNT networks were applied to macroscopically textured polydimethylsiloxane (PDMS) scaffolds. The application of CNTs to the surface of PDMS‐based scaffolds resulted in an up to 10‐fold increase in cell adherence and 240% increase in proliferation, which is attributable to increased nanoscale roughness and hydrophilicity. The introduction of macroscale features to PDMS induced alignment of chondrocytes, successfully mimicking the cell behavior observed in the superficial layer of cartilage. Raman spectroscopy was used as a noninvasive, label‐free method to monitor extracellular matrix production and chondrocyte phenotype. Chondrocytes on these scaffolds successfully produced collagen, glycosaminoglycan, and aggrecan. This study demonstrates that introducing physical features at different length scales allows for a high level of control over tissue scaffold design and, thus, cell behavior. Ultimately, these textured scaffolds can serve as platforms to improve the understanding of osteoarthritis and for early‐stage therapeutic testing.
Collapse
Affiliation(s)
- Katrín Lind Elídóttir
- Department of Physics, Faculty of Engineering and Physical Sciences, University of Surrey, Guildford, UK.,Department of Veterinary Pre-Clinical Sciences, University of Surrey, Guildford, UK
| | - Louie Scott
- Department of Veterinary Pre-Clinical Sciences, University of Surrey, Guildford, UK
| | - Rebecca Lewis
- Department of Veterinary Pre-Clinical Sciences, University of Surrey, Guildford, UK
| | - Izabela Jurewicz
- Department of Physics, Faculty of Engineering and Physical Sciences, University of Surrey, Guildford, UK
| |
Collapse
|
35
|
Castro NJ, Babakhanova G, Hu J, Athanasiou K. Nondestructive testing of native and tissue-engineered medical products: adding numbers to pictures. Trends Biotechnol 2022; 40:194-209. [PMID: 34315621 PMCID: PMC8772387 DOI: 10.1016/j.tibtech.2021.06.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 02/03/2023]
Abstract
Traditional destructive tests are used for quality assurance and control within manufacturing workflows. Their applicability to biomanufacturing is limited due to inherent constraints of the biomanufacturing process. To address this, photo- and acoustic-based nondestructive testing has risen in prominence to interrogate not only structure and function, but also to integrate quantitative measurements of biochemical composition to cross-correlate structural, compositional, and functional variances. We survey relevant literature related to single-mode and multimodal nondestructive testing of soft tissues, which adds numbers (quantitative measurements) to pictures (qualitative data). Native and tissue-engineered articular cartilage is highlighted because active biomanufacturing processes are being developed. Included are recent efforts and prominent trends focused on technologies for clinical and in-process biomanufacturing applications.
Collapse
Affiliation(s)
- Nathan J. Castro
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92617, USA
| | - Greta Babakhanova
- Biosystems and Biomaterials Division, National Institute of Standards and Technology, Gaithersburg, MD, 20899, USA
| | - Jerry Hu
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92617, USA
| | - K.A. Athanasiou
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92617, USA,Correspondence:
| |
Collapse
|
36
|
Øvrebø Ø, Perale G, Wojciechowski JP, Echalier C, Jeffers JRT, Stevens MM, Haugen HJ, Rossi F. Design and clinical application of injectable hydrogels for musculoskeletal therapy. Bioeng Transl Med 2022; 7:e10295. [PMID: 35600661 PMCID: PMC9115710 DOI: 10.1002/btm2.10295] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 11/25/2022] Open
Abstract
Musculoskeletal defects are an enormous healthcare burden and source of pain and disability for individuals. With an aging population, the proportion of individuals living with these medical indications will increase. Simultaneously, there is pressure on healthcare providers to source efficient solutions, which are cheaper and less invasive than conventional technology. This has led to an increased research focus on hydrogels as highly biocompatible biomaterials that can be delivered through minimally invasive procedures. This review will discuss how hydrogels can be designed for clinical translation, particularly in the context of the new European Medical Device Regulation (MDR). We will then do a deep dive into the clinically used hydrogel solutions that have been commercially approved or have undergone clinical trials in Europe or the United States. We will discuss the therapeutic mechanism and limitations of these products. Due to the vast application areas of hydrogels, this work focuses only on treatments of cartilage, bone, and the nucleus pulposus. Lastly, the main steps toward clinical translation of hydrogels as medical devices are outlined. We suggest a framework for how academics can assist small and medium MedTech enterprises conducting the initial clinical investigation and post‐market clinical follow‐up required in the MDR. It is evident that the successful translation of hydrogels is governed by acquiring high‐quality pre‐clinical and clinical data confirming the device mechanism of action and safety.
Collapse
Affiliation(s)
- Øystein Øvrebø
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoMilanoItaly
- Department of BiomaterialsInstitute of Clinical Dentistry, University of OsloOsloNorway
- Material Biomimetic ASOslo Science ParkOsloNorway
| | - Giuseppe Perale
- Industrie Biomediche Insubri SAMezzovico‐ViraSwitzerland
- Faculty of Biomedical SciencesUniversity of Southern SwitzerlandLuganoSwitzerland
- Ludwig Boltzmann Institute for Experimental and Clinical TraumatologyViennaAustria
| | - Jonathan P. Wojciechowski
- Department of MaterialsImperial College LondonLondonUK
- Department of BioengineeringImperial College LondonLondonUK
- Institute of Biomedical EngineeringImperial College LondonLondonUK
| | - Cécile Echalier
- Department of MaterialsImperial College LondonLondonUK
- Department of BioengineeringImperial College LondonLondonUK
- Institute of Biomedical EngineeringImperial College LondonLondonUK
- Hybrid Technology Hub, Centre of ExcellenceInstitute of Basic Medical Science, University of OsloOsloNorway
| | | | - Molly M. Stevens
- Department of MaterialsImperial College LondonLondonUK
- Department of BioengineeringImperial College LondonLondonUK
- Institute of Biomedical EngineeringImperial College LondonLondonUK
| | - Håvard J. Haugen
- Department of BiomaterialsInstitute of Clinical Dentistry, University of OsloOsloNorway
- Material Biomimetic ASOslo Science ParkOsloNorway
| | - Filippo Rossi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoMilanoItaly
| |
Collapse
|
37
|
Pezzotti G, Zhu W, Terai Y, Marin E, Boschetto F, Kawamoto K, Itaka K. Raman spectroscopic insight into osteoarthritic cartilage regeneration by mRNA therapeutics encoding cartilage-anabolic transcription factor Runx1. Mater Today Bio 2022; 13:100210. [PMID: 35281370 PMCID: PMC8913780 DOI: 10.1016/j.mtbio.2022.100210] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/18/2022] [Accepted: 01/28/2022] [Indexed: 11/05/2022] Open
Abstract
While joint arthroplasty remains nowadays the most popular option available to repair chronically degenerated osteoarthritic joints, possibilities are recently emerging for regeneration of damaged cartilage rather than its replacement with artificial biomaterials. This latter strategy could allow avoiding the quite intrusive surgical procedures associated with total joint replacement. Building upon this notion, we first apply Raman spectroscopy to characterize diseased cartilage in a mice model of instability-induced knee osteoarthritis (OA) upon medial collateral ligament (MCL) and medial meniscus (MM) transections. Then, we examine the same OA model after cartilage regeneration by means of messenger RNA (mRNA) delivery of a cartilage-anabolic runt-related transcription factor 1 (RUNX1). Raman spectroscopy is shown to substantiate at the molecular scale the therapeutic effect of the Runx1 mRNA cartilage regeneration approach. This study demonstrates how the Raman spectroscopic method could support and accelerate the development of new therapies for cartilage diseases.
Collapse
|
38
|
Brown ETT, Simons JMLJW, Thambyah A. The ultrastructure of cartilage tissue and its swelling response in relation to matrix health. J Anat 2022; 240:107-119. [PMID: 34333796 PMCID: PMC8655166 DOI: 10.1111/joa.13527] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 06/24/2021] [Accepted: 07/20/2021] [Indexed: 11/26/2022] Open
Abstract
This multi-length scale anatomical study explores the influence of mild cartilage structural degeneration on the tissue swelling response. While the swelling response of cartilage has been studied extensively, this is the first study to reveal and correlate tissue microstructure and ultrastructure, with the swelling induced cartilage tissue strains. Cartilage sample strips (n = 30) were obtained from the distal-lateral quadrant of thirty mildly degenerate bovine patellae and, following excision from the bone, the cartilage strips were allowed to swell freely for 2 h in solutions of physiological saline and distilled water successively. The swelling response of this group of samples were compared with that of healthy cartilage, with (n = 20) and without the surface layer (n = 20). The subsequent curling response of cartilage showed that in healthy tissue it was highly variable, and with the surface removed some samples curved in the opposite direction, while in the mildly degenerate tissue group, virtually all tissue strips curved in a consistent upward manner. A significant difference in strain was observed between healthy samples with surface layer removed and mildly degenerate samples, illustrating how excision of the surface zone from pristine cartilage is insufficient to model the swelling response of tissue which has undergone natural degenerative changes. On average, total tissue thickness increased from 940 µm (healthy) to 1079 µm (mildly degenerate), however, looking at the zonal strata, surface and transition zone thicknesses both decreased while deep zone thickness increased from healthy to mildly degenerate tissue. Morphologically, changes to the surface zone integrity were correlated with a diminished surface layer which, at the ultrastructural scale, correlated with a decreased fibrillar density. Similarly, fibrosity of the general matrix visible at the microscale was associated with a loss of later interconnectivity resulting in large, aggregated fibril bundles. The microstructural and ultrastructural investigation revealed that the key differences influencing the tissue swelling strain response was (1) the thickness and extent of disruption to the surface layer and (2) the amount of fibrillar network destructuring, highlighting the importance of the collagen and tissue matrix structure in restraining cartilage swelling.
Collapse
Affiliation(s)
- Emma Te Tūmanako Brown
- Department of Chemical and Materials EngineeringUniversity of AucklandAucklandNew Zealand
| | - Joni M. L. J. W. Simons
- Orthopaedic Biomechanics GroupDepartment of Biomedical EngineeringEindhoven University of TechnologyEindhovenThe Netherlands
| | - Ashvin Thambyah
- Department of Chemical and Materials EngineeringUniversity of AucklandAucklandNew Zealand
| |
Collapse
|
39
|
Querido W, Zouaghi S, Padalkar M, Morman J, Falcon J, Kandel S, Pleshko N. Nondestructive assessment of tissue engineered cartilage based on biochemical markers in cell culture media: application of attenuated total reflection Fourier transform infrared (ATR-FTIR) spectroscopy. Analyst 2022; 147:1730-1741. [PMID: 35343541 PMCID: PMC9047556 DOI: 10.1039/d1an02351a] [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
ATR spectral data obtained from cell culture medium discards can be used to assess glucose and lactate content, which are shown here to be a surrogate for matrix development in tissue engineered cartilage.
Collapse
Affiliation(s)
- William Querido
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, USA
| | - Sabrina Zouaghi
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, USA
| | - Mugdha Padalkar
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, USA
| | - Justin Morman
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, USA
| | - Jessica Falcon
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, USA
| | - Shital Kandel
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, USA
| | - Nancy Pleshko
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, USA
| |
Collapse
|
40
|
Cialla-May D, Krafft C, Rösch P, Deckert-Gaudig T, Frosch T, Jahn IJ, Pahlow S, Stiebing C, Meyer-Zedler T, Bocklitz T, Schie I, Deckert V, Popp J. Raman Spectroscopy and Imaging in Bioanalytics. Anal Chem 2021; 94:86-119. [PMID: 34920669 DOI: 10.1021/acs.analchem.1c03235] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Dana Cialla-May
- Leibniz-Institute of Photonic Technology, Member of the Leibniz Research Alliance - Leibniz Health Technologies, Albert-Einstein-Str. 9, 07745 Jena, Germany.,Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University, Helmholtzweg 4, 07743 Jena, Germany.,InfectoGnostics Research Campus Jena, Center of Applied Research, Philosophenweg 7, 07743 Jena, Germany
| | - Christoph Krafft
- Leibniz-Institute of Photonic Technology, Member of the Leibniz Research Alliance - Leibniz Health Technologies, Albert-Einstein-Str. 9, 07745 Jena, Germany
| | - Petra Rösch
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University, Helmholtzweg 4, 07743 Jena, Germany
| | - Tanja Deckert-Gaudig
- Leibniz-Institute of Photonic Technology, Member of the Leibniz Research Alliance - Leibniz Health Technologies, Albert-Einstein-Str. 9, 07745 Jena, Germany.,Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University, Helmholtzweg 4, 07743 Jena, Germany
| | - Torsten Frosch
- Leibniz-Institute of Photonic Technology, Member of the Leibniz Research Alliance - Leibniz Health Technologies, Albert-Einstein-Str. 9, 07745 Jena, Germany.,Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University, Helmholtzweg 4, 07743 Jena, Germany
| | - Izabella J Jahn
- Leibniz-Institute of Photonic Technology, Member of the Leibniz Research Alliance - Leibniz Health Technologies, Albert-Einstein-Str. 9, 07745 Jena, Germany.,Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University, Helmholtzweg 4, 07743 Jena, Germany
| | - Susanne Pahlow
- Leibniz-Institute of Photonic Technology, Member of the Leibniz Research Alliance - Leibniz Health Technologies, Albert-Einstein-Str. 9, 07745 Jena, Germany.,Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University, Helmholtzweg 4, 07743 Jena, Germany.,InfectoGnostics Research Campus Jena, Center of Applied Research, Philosophenweg 7, 07743 Jena, Germany
| | - Clara Stiebing
- Leibniz-Institute of Photonic Technology, Member of the Leibniz Research Alliance - Leibniz Health Technologies, Albert-Einstein-Str. 9, 07745 Jena, Germany
| | - Tobias Meyer-Zedler
- Leibniz-Institute of Photonic Technology, Member of the Leibniz Research Alliance - Leibniz Health Technologies, Albert-Einstein-Str. 9, 07745 Jena, Germany.,Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University, Helmholtzweg 4, 07743 Jena, Germany
| | - Thomas Bocklitz
- Leibniz-Institute of Photonic Technology, Member of the Leibniz Research Alliance - Leibniz Health Technologies, Albert-Einstein-Str. 9, 07745 Jena, Germany.,Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University, Helmholtzweg 4, 07743 Jena, Germany
| | - Iwan Schie
- Leibniz-Institute of Photonic Technology, Member of the Leibniz Research Alliance - Leibniz Health Technologies, Albert-Einstein-Str. 9, 07745 Jena, Germany.,Ernst-Abbe-Hochschule Jena, University of Applied Sciences, Department of Biomedical Engineering and Biotechnology, Carl-Zeiss-Promenade 2, 07745 Jena, Germany
| | - Volker Deckert
- Leibniz-Institute of Photonic Technology, Member of the Leibniz Research Alliance - Leibniz Health Technologies, Albert-Einstein-Str. 9, 07745 Jena, Germany.,Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University, Helmholtzweg 4, 07743 Jena, Germany
| | - Jürgen Popp
- Leibniz-Institute of Photonic Technology, Member of the Leibniz Research Alliance - Leibniz Health Technologies, Albert-Einstein-Str. 9, 07745 Jena, Germany.,Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University, Helmholtzweg 4, 07743 Jena, Germany.,InfectoGnostics Research Campus Jena, Center of Applied Research, Philosophenweg 7, 07743 Jena, Germany
| |
Collapse
|
41
|
Zhou H, Piñeiro Llanes J, Sarntinoranont M, Subhash G, Simmons CS. Label-free quantification of soft tissue alignment by polarized Raman spectroscopy. Acta Biomater 2021; 136:363-374. [PMID: 34537413 DOI: 10.1016/j.actbio.2021.09.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/24/2021] [Accepted: 09/09/2021] [Indexed: 11/29/2022]
Abstract
The organization of proteins is an important determinant of functionality in soft tissues. However, such organization is difficult to monitor over time in soft tissue with complex compositions. Here, we establish a method to determine the alignment of proteins in soft tissues of varying composition by polarized Raman spectroscopy (PRS). Unlike most conventional microscopy methods, PRS leverages non-destructive, label-free sample preparation. PRS data from highly aligned muscle layers were utilized to derive a weighting function for aligned proteins via principal component analysis (PCA). This trained weighting function was used as a master loading function to calculate a principal component score (PC1 Score) as a function of polarized angle for tendon, dermis, hypodermis, and fabricated collagen gels. Since the PC1 Score calculated at arbitrary angles was insufficient to determine level of alignment, we developed an Amplitude Alignment Metric by fitting a sine function to PC1 Score with respect to polarized angle. We found that our PRS-based Amplitude Alignment Metric can be used as an indicator of level of protein alignment in soft tissues in a non-destructive manner with label-free preparation and has similar discriminatory capacity among isotropic and anisotropic samples compared to microscopy-based image processing method. This PRS method does not require a priori knowledge of sample orientation nor composition and appears insensitive to changes in protein composition among different tissues. The Amplitude Alignment Metric introduced here could enable convenient and adaptable evaluation of protein alignment in soft tissues of varying protein and cell composition. STATEMENT OF SIGNIFICANCE: Polarized Raman spectroscopy (PRS) has been used to characterize the of organization of soft tissues. However, most of the reported applications of PRS have been on collagen-rich tissues and reliant on intensities of collagen-related vibrations. This work describes a PRS method via a multivariate analysis to characterize alignment in soft tissues composed of varying proteins. Of note, the highly aligned muscle layer of mouse skin was used to train a master function then applied to other soft tissue samples, and the degree of anisotropy in the PRS response was evaluated to obtain the level of alignment in tissues. We have demonstrated that this method supports convenient and adaptable evaluation of protein alignment in soft tissues of varying protein and cell composition.
Collapse
Affiliation(s)
- Hui Zhou
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida, USA
| | - Janny Piñeiro Llanes
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida, USA
| | - Malisa Sarntinoranont
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida, USA
| | - Ghatu Subhash
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida, USA
| | - Chelsey S Simmons
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida, USA.
| |
Collapse
|
42
|
Sherlock B, Chen J, Mansfield J, Green E, Winlove C. Biophotonic tools for probing extracellular matrix mechanics. Matrix Biol Plus 2021; 12:100093. [PMID: 34934939 PMCID: PMC8661043 DOI: 10.1016/j.mbplus.2021.100093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 11/07/2021] [Accepted: 11/11/2021] [Indexed: 12/11/2022] Open
Abstract
The complex, hierarchical and heterogeneous biomechanics of the extracellular matrix (ECM) are central to the health of multicellular organisms. Characterising the distribution, dynamics and above all else origins of ECM biomechanics are challenges that have captivated researchers for decades. Recently, a suite of biophotonics techniques have emerged as powerful new tools to investigate ECM biomechanics. In this mini-review, we discuss how the non-destructive, sub-micron resolution imaging capabilities of Raman spectroscopy and nonlinear microscopy are being used to interrogate the biomechanics of thick, living tissues. These high speed, label-free techniques are implemented during mechanical testing, providing unprecedented insight into the compositional and structural response of the ECM to changes in the mechanical environment.
Collapse
Affiliation(s)
- B.E. Sherlock
- College of Medicine and Health, University of Exeter, Exeter EX1 2LU, United Kingdom
| | - J. Chen
- College of Engineering, Mathematical and Physical Sciences, University of Exeter, Exeter EX4 4QF, United Kingdom
| | - J.C. Mansfield
- College of Engineering, Mathematical and Physical Sciences, University of Exeter, Exeter EX4 4QF, United Kingdom
| | - E. Green
- College of Engineering, Mathematical and Physical Sciences, University of Exeter, Exeter EX4 4QF, United Kingdom
| | - C.P. Winlove
- College of Engineering, Mathematical and Physical Sciences, University of Exeter, Exeter EX4 4QF, United Kingdom
| |
Collapse
|
43
|
Gao T, Boys AJ, Zhao C, Chan K, Estroff LA, Bonassar LJ. Non-Destructive Spatial Mapping of Glycosaminoglycan Loss in Native and Degraded Articular Cartilage Using Confocal Raman Microspectroscopy. Front Bioeng Biotechnol 2021; 9:744197. [PMID: 34778225 PMCID: PMC8581176 DOI: 10.3389/fbioe.2021.744197] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/24/2021] [Indexed: 12/02/2022] Open
Abstract
Articular cartilage is a collagen-rich tissue that provides a smooth, lubricated surface for joints and is also responsible for load bearing during movements. The major components of cartilage are water, collagen, and proteoglycans. Osteoarthritis is a degenerative disease of articular cartilage, in which an early-stage indicator is the loss of proteoglycans from the collagen matrix. In this study, confocal Raman microspectroscopy was applied to study the degradation of articular cartilage, specifically focused on spatially mapping the loss of glycosaminoglycans (GAGs). Trypsin digestion was used as a model for cartilage degradation. Two different scanning geometries for confocal Raman mapping, cross-sectional and depth scans, were applied. The chondroitin sulfate coefficient maps derived from Raman spectra provide spatial distributions similar to histological staining for glycosaminoglycans. The depth scans, during which subsurface data were collected without sectioning the samples, can also generate spectra and GAG distributions consistent with Raman scans of the surface-to-bone cross sections. In native tissue, both scanning geometries demonstrated higher GAG content at the deeper zone beneath the articular surface and negligible GAG content after trypsin degradation. On partially digested samples, both scanning geometries detected an ∼100 μm layer of GAG depletion. Overall, this research provides a technique with high spatial resolution (25 μm pixel size) to measure cartilage degradation without tissue sections using confocal Raman microspectroscopy, laying a foundation for potential in vivo measurements and osteoarthritis diagnosis.
Collapse
Affiliation(s)
- Tianyu Gao
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, United States
| | - Alexander J Boys
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, United States
| | - Crystal Zhao
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, United States
| | - Kiara Chan
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, United States
| | - Lara A Estroff
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, United States.,Kavli Institute at Cornell for Nanoscale Science, Ithaca, NY, United States
| | - Lawrence J Bonassar
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, United States.,Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| |
Collapse
|
44
|
Collagen orientation probed by polarized Raman spectra can serve as differential diagnosis indicator between different grades of meniscus degeneration. Sci Rep 2021; 11:20299. [PMID: 34645874 PMCID: PMC8514572 DOI: 10.1038/s41598-021-99569-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/19/2021] [Indexed: 11/08/2022] Open
Abstract
The purpose of the present study was to analyze normal and degenerated menisci with Raman methodology on thin sections of formalin fixed paraffin embedding tissues and to correlate the Raman findings with the grade of meniscus degeneration. Menisci (n = 27) were removed from human knee joints after total knee replacement or meniscectomy. Following routine histopathological analysis to determine the grade of meniscal lesions obtained from healthy and degenerated formaline fixed paraffin embedded (FFPE) meniscal sections, Raman polarization approach was applied to evaluate the orientation of collagen fibrils in different levels of the same 5 μm thick FFPE meniscal tissue sections, used for histopathological assessment. We collected Raman spectra in two different polarization geometries, v-HH and v-VV, and calculated the mean value of the v-HH/v-VV intensity ratio of two Raman bands, sensitive and non-sensitive to the molecular orientation. The collagen specific amide I band at 1665 cm-1, has the higher sensitivity dependence on the Raman polarization. The mean values of ratio v-HH/v-VV of the 1665 cm-1 peak intensity was significantly higher in healthy, mean ± SD: 2.56 ± 0.46, compared to degenerated menisci, mean ± SD: 1.85 ± 0.42 (p = 0.0014). The mean values of v-HH/v-VV intensity ratio were 2.18 and 1.50 for low and high degenerated menisci, respectively (p < 0.0001). The difference of peak intensities in the two laser polarizations is decreased in the degenerated meniscus; this difference is diminishing as the degeneration increases. The v-HH/v-VV ratio was also of significant difference in low as compared to control and high grade meniscus lesions (p = 0.036 and p < 0.0001, respectively) offering valuable information for the approach of its biology and function. In the present study we showed that the 5 μm thick sections can be used for Raman analysis of meniscal tissue with great reliability, in terms of sensitivity, specificity, false-negative and false-positive results. Our data introduce the interesting hypothesis that compact portable Raman microscopy on tissue sections can be used intra-operatively for fast diagnosis and hence, accurate procedure design in the operating room.
Collapse
|
45
|
Bharadwaz A, Jayasuriya AC. Fabrication of porous chitosan particles using a novel two-step porogen leaching and lyophilization method with the label-free multivariate spectral assessment of live adhered cells. Colloids Surf B Biointerfaces 2021; 208:112094. [PMID: 34500203 DOI: 10.1016/j.colsurfb.2021.112094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 01/15/2023]
Abstract
Porous chitosan (CS) particles were fabricated using a novel two-step technique that employed a porogen leaching phase followed by lyophilization or freeze-drying. Poly(ethylene glycol) (PEG) was mixed as a porogen in two different quantities with the CS solution before particle synthesis via coacervation. After the PEG leached out into deionized (DI) water at an elevated constant temperature, the final freeze-dried CS particles revealed surface features that resembled pore pockets. A three-dimensional (3D) culture of murine osteoblast cell line (OB-6) was seeded on these particles to analyze the effect of the porous structure on the cell activity, as compared to a control group with no added porogen. The results highlighted an enhancement in cell adhesion and proliferation on the two porous sample groups. A Raman spectroscopy-based label-free technique for live cell biomarker analysis was applied using multivariate spectral analysis. Results of the spectral analysis in the molecular fingerprint region corresponding to the Raman shift between 900 cm-1 and 1700 cm-1inferred inter-group variations. The bands at 1005 cm-1 and 1375 cm-1 were assigned to the live cell biomarkers phenylalanine and glycosaminoglycan, respectively, and were assessed during the multivariate spectral analysis. The corresponding score plot and loading information generated from the Principal Component Analysis (PCA) of the Raman spectrum at day 7 and day 14, pointed at inter-group spectral variations related to cell adhesion and proliferation between the two porous CS particle groups and the control.
Collapse
Affiliation(s)
- Angshuman Bharadwaz
- Biomedical Engineering Program, Department of Bioengineering, College of Engineering, The University of Toledo, Toledo, OH, 43606, USA
| | - Ambalangodage C Jayasuriya
- Biomedical Engineering Program, Department of Bioengineering, College of Engineering, The University of Toledo, Toledo, OH, 43606, USA; Department of Orthopaedic Surgery, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA.
| |
Collapse
|
46
|
Sistani MN, Zavareh S, Valujerdi MR, Salehnia M. Characteristics of a decellularized human ovarian tissue created by combined protocols and its interaction with human endometrial mesenchymal cells. Prog Biomater 2021; 10:195-206. [PMID: 34482521 DOI: 10.1007/s40204-021-00163-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/30/2021] [Indexed: 01/15/2023] Open
Abstract
The present study makes assessments by analyzing the efficacy of combined decellularization protocol for human ovarian fragments. Tissues were decellularized by freeze-thaw cycles, and treated with Triton X-100 and four concentrations (0.1, 0.5, 1 and 1.5%) of sodium dodecyl sulfate (SDS) at two exposure times. The morphology and DNA content of decellularized tissues were analyzed, and the group with better morphology and lower DNA content was selected for further assessments. The Acridine orange, Masson's trichrome, Alcian blue, and Periodic Acid-Schiff staining were used for extracellular matrix (ECM) evaluation. The amount of collagen types I and IV, glycosaminoglycans (GAGs), and elastin was quantified by Raman spectroscopy. The fine structure of the scaffold by scanning electron microscopy was studied. The endometrial mesenchymal cells were seeded onto decellularized scaffold by centrifugal method and cultured for 7 days. After 72 h the treated group with 0.5% SDS showed well-preserved ECM morphology with the minimum level of DNA (2.23% ± 0.08). Raman spectroscopy analysis confirmed that, the amount of ECM components was not significantly decreased in the decellularized group (P < 0.001) in comparison with native control. The electron micrographs demonstrated that the porosity and structure of ECM fibers in the decellularized group was similar to native ovary. The endometrial mesenchymal cells were attached and penetrated into the decellularized scaffold. In conclusion this combined protocol was an effective method to decellularize human ovarian tissue with high preservation of ECM contents, and human endometrial mesenchymal cells which successfully interacted with this created scaffold.
Collapse
Affiliation(s)
- Maryam Nezhad Sistani
- Anatomy Department, Faculty of Medical Sciences, Tarbiat Modares University, P.O. BOX: 14115-111, Tehran, Iran
| | - Saeed Zavareh
- School of Biology, Damghan University, Damghan, Iran
| | - Mojtaba Rezazadeh Valujerdi
- Anatomy Department, Faculty of Medical Sciences, Tarbiat Modares University, P.O. BOX: 14115-111, Tehran, Iran
| | - Mojdeh Salehnia
- Anatomy Department, Faculty of Medical Sciences, Tarbiat Modares University, P.O. BOX: 14115-111, Tehran, Iran.
| |
Collapse
|
47
|
Cui H, Glidle A, Cooper JM. Spatial Heterodyne Offset Raman Spectroscopy Enabling Rapid, High Sensitivity Characterization of Materials' Interfaces. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2101114. [PMID: 34013665 DOI: 10.1002/smll.202101114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/18/2021] [Indexed: 06/12/2023]
Abstract
Spatially offset Raman spectroscopy is integrated with a fiber-coupled spatial heterodyne spectrometer to collect Raman spectra from deep within opaque or scattering materials. The method, named spatial heterodyne offset Raman spectroscopy generates a wavenumber-dependent spatial phase shift of the optical signal as a "spectral" image on a charge-coupled device detector. The image can be readily processed from the spatial domain using a single, simple, and "on-the-fly" Fourier transform to generate Raman spectra, in the frequency domain. By collecting all of the spatially offset Raman scattered photons that pass through the microscope's collection objective lens, the methodology gives an improvement in the Raman sensitivity by an order of magnitude. The instrumentation is both mechanically robust and "movement-free," which when coupled with the associated advantages of highly efficient signal collection and ease of data processing, enables rapid interfacial analysis of complex constructs based on established biomaterials models.
Collapse
Affiliation(s)
- Han Cui
- Beijing Key Lab for Precision Optoelectronic Measurement Instrument and Technology, School of Optics and Photonics, Beijing Institute of Technology, Beijing, 100081, China
- Division of Biomedical Engineering, James Watt School of Engineering, University of Glasgow, Glasgow, G12 8LT, UK
| | - Andrew Glidle
- Division of Biomedical Engineering, James Watt School of Engineering, University of Glasgow, Glasgow, G12 8LT, UK
| | - Jonathan M Cooper
- Division of Biomedical Engineering, James Watt School of Engineering, University of Glasgow, Glasgow, G12 8LT, UK
| |
Collapse
|
48
|
Cao Y, Cheng P, Sang S, Xiang C, An Y, Wei X, Shen Z, Zhang Y, Li P. Mesenchymal stem cells loaded on 3D-printed gradient poly(ε-caprolactone)/methacrylated alginate composite scaffolds for cartilage tissue engineering. Regen Biomater 2021; 8:rbab019. [PMID: 34211731 PMCID: PMC8240606 DOI: 10.1093/rb/rbab019] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/08/2021] [Accepted: 04/12/2021] [Indexed: 01/06/2023] Open
Abstract
Cartilage has limited self-repair ability due to its avascular, alymphatic and aneural features. The combination of three-dimensional (3D) printing and tissue engineering provides an up-and-coming approach to address this issue. Here, we designed and fabricated a tri-layered (superficial layer (SL), middle layer (ML) and deep layer (DL)) stratified scaffold, inspired by the architecture of collagen fibers in native cartilage tissue. The scaffold was composed of 3D printed depth-dependent gradient poly(ε-caprolactone) (PCL) impregnated with methacrylated alginate (ALMA), and its morphological analysis and mechanical properties were tested. To prove the feasibility of the composite scaffolds for cartilage regeneration, the viability, proliferation, collagen deposition and chondrogenic differentiation of embedded rat bone marrow mesenchymal stem cells (BMSCs) in the scaffolds were assessed by Live/dead assay, CCK-8, DNA content, cell morphology, immunofluorescence and real-time reverse transcription polymerase chain reaction. BMSCs-loaded gradient PCL/ALMA scaffolds showed excellent cell survival, cell proliferation, cell morphology, collagen II deposition and hopeful chondrogenic differentiation compared with three individual-layer scaffolds. Hence, our study demonstrates the potential use of the gradient PCL/ALMA construct for enhanced cartilage tissue engineering.
Collapse
Affiliation(s)
- Yanyan Cao
- Key Lab of Advanced Transducers and Intelligent Control System of the Ministry of Education, MicroNano System Research Center, College of Information and Computer, Taiyuan University of Technology, Taiyuan 030024, China.,College of Information Science and Engineering, Hebei North University, Zhangjiakou 075000, China
| | - Peng Cheng
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Shengbo Sang
- Key Lab of Advanced Transducers and Intelligent Control System of the Ministry of Education, MicroNano System Research Center, College of Information and Computer, Taiyuan University of Technology, Taiyuan 030024, China
| | - Chuan Xiang
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Yang An
- Department of Plastic Surgery, Peking University Third Hospital, Beijing 100191, China
| | - Xiaochun Wei
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Zhizhong Shen
- Key Lab of Advanced Transducers and Intelligent Control System of the Ministry of Education, MicroNano System Research Center, College of Information and Computer, Taiyuan University of Technology, Taiyuan 030024, China
| | - Yixia Zhang
- Department of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Pengcui Li
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
49
|
Han L, Xu N, Lv S, Yin J, Zheng D, Li X. Enhanced In Vitro and In Vivo Efficacy of Alginate/Silk Protein/Hyaluronic Acid with Polypeptide Microsphere Delivery for Tissue Regeneration of Articular Cartilage. J Biomed Nanotechnol 2021; 17:901-909. [PMID: 34082875 DOI: 10.1166/jbn.2021.3071] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Alginate/Silk fibroin/hyaluronic acid (ALG/SF/HA) nanocomposites were synthesised using blending, inter-linking, and lyophilization methods. We investigated the physicochemical properties of the resulting nanocomposites, including their water retention, weight loss, porosity and cytocompatibility. The optimum ratios of the ALG/SF/HA scaffolding were 3:6.5:0.5. Nanocomposites with optimum ratios were then prepared by integrating pilose antler polypeptides (PAPS) to poly(lactic-co-glycolic acid) (PLGA) microspheres, and the performance was investigated. PAPS-ALG/SF/HA nanocomposites exhibited desirable adhesions and proliferations. Rabbit cartilage deficiencies was developed by the animal model. The cartilage repair effects deficiencies were detected and analyzed between PAPS-SF/ALG/ALG/SF/HA, and control activity classes. The deficiencies were virtually fully remedied after 13 weeks in the presence of PAPS-ALG/SF/HA class, suggesting that the PAPS-ALG/SF/HA nanocomposites had a positive effects on joint cartilage repair.
Collapse
Affiliation(s)
- Long Han
- Department of Orthopedics, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213003, China
| | - Nanwei Xu
- Department of Orthopedics, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213003, China
| | - Songwei Lv
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Jianjian Yin
- Department of Orthopedics, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213003, China
| | - Dong Zheng
- Department of Orthopedics, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213003, China
| | - Xin Li
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| |
Collapse
|
50
|
Raman microspectroscopy and Raman imaging reveal biomarkers specific for thoracic aortic aneurysms. CELL REPORTS MEDICINE 2021; 2:100261. [PMID: 34095874 PMCID: PMC8149374 DOI: 10.1016/j.xcrm.2021.100261] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 01/29/2021] [Accepted: 04/06/2021] [Indexed: 01/30/2023]
Abstract
Aortic rupture and dissection are life-threatening complications of ascending thoracic aortic aneurysms (aTAAs), and risk assessment has been largely based on the monitoring of lumen size enlargement. Temporal changes in the extracellular matrix (ECM), which has a critical impact on aortic remodeling, are not routinely evaluated, and cardiovascular biomarkers do not exist to predict aTAA formation. Here, Raman microspectroscopy and Raman imaging are used to identify spectral biomarkers specific for aTAAs in mice and humans by multivariate data analysis (MVA). Multivariate curve resolution-alternating least-squares (MCR-ALS) combined with Lasso regression reveals elastic fiber-derived (Ce1) and collagen fiber-derived (Cc6) components that are significantly increased in aTAA lesions of murine and human aortic tissues. In particular, Cc6 detects changes in amino acid residues, including phenylalanine, tyrosine, tryptophan, cysteine, aspartate, and glutamate. Ce1 and Cc6 may serve as diagnostic Raman biomarkers that detect alterations of amino acids derived from aneurysm lesions. Label-free Raman imaging of human/murine ascending thoracic aortic aneurysm (aTAA) Multivariate analysis of Raman spectra allows detection of aTAA molecular features Identification of spectral biomarkers for aTAA in elastic and collagen fibers Alterations in amino acid spectra correlate with aTAA formation
Collapse
|