Multifunctional Scaffolds for Musculoskeletal Regeneration

A special issue of Bioengineering (ISSN 2306-5354). This special issue belongs to the section "Regenerative Engineering".

Deadline for manuscript submissions: closed (30 September 2022) | Viewed by 63236

Special Issue Editors

Department of Restorative Science, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA
Interests: biomaterials; nanomaterials; surface modification; material characterisation; tissue engineering and regenerative medicine
Special Issues, Collections and Topics in MDPI journals
Bone & Joint Research Group, University of Southampton, Southampton, UK
Interests: biomaterials; drug delivery system; immuomodulation; tissue regeneration
School of Dentistry, University of Minnesota, Twin Cities, Minneapolis, Minneapolis, MN, USA
Interests: surface modification for dental and orthopedic implants; biomineralization of synthetic materials and tissues; biomimetic systems for biomedical applications; development and evaluation of dental restorative materials; regeneration of hard tissues (enamel, dentin, bone); dental biofilms; synthetic peptides, recombinant biopolymers, and self-assembly

Special Issue Information

Dear Colleagues,

With continuously rising incidences of musculoskeletal-related injuries and disease that require surgical intervention, a considerable and increasing number of patients are experiencing pain and loss of mobility. This debilitating condition has huge socio-economic burden, with direct implications for the patient and the healthcare system. Current treatment modalities include the use of autografts, allografts, or non-tissue prosthetics; however, they have limited success due to challenged availability, donor site morbidity, tissue rejection due to immune incompatibility, incomplete tissue regeneration, or infection at the surgical site. This has prompted the need for designing bioactive scaffolds with or without cell therapy as tissue replacement alternatives. Bioactive scaffolds are not only biocompatible, but they elicit targeted biological responses so that tissue constructs can seemingly integrate with the surrounding tissue. Significant advancements have been made in designing bioactive scaffolds, from matching the mechanical properties of the tissue of interest to combining strategies for the controlled time-dependent delivery of growth factors to emulate the events involved in tissue regeneration. The field has seen a drift towards the incorporation of additional bioactive agents to accomplish a plethora of different functions, such as antibacterial compounds to combat infection or immunomodulating factors to augment tissue repair.

This Special Issue is envisaged to present the state of the art of basic, translational, and clinical efforts that combine these different strategies to design and develop bioactive scaffolds with multifaceted capabilities. Different topics of interest to this Special Issue include: 1) surface modification strategies to support cell migration, cell homing, and tissue regeneration; 2) designing immunomodulatory scaffolds for creating a reparative environment conducive to tissue regeneration; 3) combining scaffolds with biologics (growth factors, peptides, small molecules) and/or inorganic fillers for improved cellular response; and 4) multifunctional antimicrobial and cell-instructive scaffolds for musculoskeletal repair.

Dr. Isha Mutreja
Dr. Yanghee Kim
Prof. Conrado Aparicio
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Bioengineering is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • multifunctional scaffolds
  • bioactive scaffolds
  • nanocomposites
  • bioinspiration
  • surface modification growth factors
  • immunomodulation
  • antibacterial
  • musculoskeletal repair and/or regeneration
  • drug delivery systems

Published Papers (17 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

14 pages, 5830 KiB  
Article
Engineering Skeletal Muscle Grafts with PAX7::GFP-Sorted Human Pluripotent Stem Cell-Derived Myogenic Progenitors on Fibrin Microfiber Bundles for Tissue Regeneration
by Sarah M. Somers, Jordana Gilbert-Honick, In Young Choi, Emily K. W. Lo, HoTae Lim, Shaquielle Dias, Kathryn R. Wagner, Hai-Quan Mao, Patrick Cahan, Gabsang Lee and Warren L. Grayson
Bioengineering 2022, 9(11), 693; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering9110693 - 15 Nov 2022
Cited by 2 | Viewed by 1823
Abstract
Tissue engineering strategies that combine human pluripotent stem cell-derived myogenic progenitors (hPDMs) with advanced biomaterials provide promising tools for engineering 3D skeletal muscle grafts to model tissue development in vitro and promote muscle regeneration in vivo. We recently demonstrated (i) the potential for [...] Read more.
Tissue engineering strategies that combine human pluripotent stem cell-derived myogenic progenitors (hPDMs) with advanced biomaterials provide promising tools for engineering 3D skeletal muscle grafts to model tissue development in vitro and promote muscle regeneration in vivo. We recently demonstrated (i) the potential for obtaining large numbers of hPDMs using a combination of two small molecules without the overexpression of transgenes and (ii) the application of electrospun fibrin microfiber bundles for functional skeletal muscle restoration following volumetric muscle loss. In this study, we aimed to demonstrate that the biophysical cues provided by the fibrin microfiber bundles induce hPDMs to form engineered human skeletal muscle grafts containing multinucleated myotubes that express desmin and myosin heavy chains and that these grafts could promote regeneration following skeletal muscle injuries. We tested a genetic PAX7 reporter line (PAX7::GFP) to sort for more homogenous populations of hPDMs. RNA sequencing and gene set enrichment analyses confirmed that PAX7::GFP-sorted hPDMs exhibited high expression of myogenic genes. We tested engineered human skeletal muscle grafts derived from PAX7::GFP-sorted hPDMs within in vivo skeletal muscle defects by assessing myogenesis, engraftment and immunogenicity using immunohistochemical staining. The PAX7::GFP-sorted groups had moderately high vascular infiltration and more implanted cell association with embryonic myosin heavy chain (eMHC) regions, suggesting they induced pro-regenerative microenvironments. These findings demonstrated the promise for the use of PAX7::GFP-sorted hPDMs on fibrin microfiber bundles and provided some insights for improving the cell–biomaterial system to stimulate more robust in vivo skeletal muscle regeneration. Full article
(This article belongs to the Special Issue Multifunctional Scaffolds for Musculoskeletal Regeneration)
Show Figures

Figure 1

13 pages, 22352 KiB  
Article
Biocompatible Nanocomposite Coatings Deposited via Layer-by-Layer Assembly for the Mechanical Reinforcement of Highly Porous Interconnected Tissue-Engineered Scaffolds
by Aoife McFerran, Mary Josephine McIvor, Patrick Lemoine, Brian J. Meenan and Jonathan G. Acheson
Bioengineering 2022, 9(10), 585; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering9100585 - 20 Oct 2022
Cited by 5 | Viewed by 1582
Abstract
Tissue-engineered (TE) scaffolds provide an ‘off-the-shelf’ alternative to autograft procedures and can potentially address their associated complications and limitations. The properties of TE scaffolds do not always match the surrounding bone, often sacrificing porosity for improved compressive strength. Previously, the layer-by-layer (LbL) assembly [...] Read more.
Tissue-engineered (TE) scaffolds provide an ‘off-the-shelf’ alternative to autograft procedures and can potentially address their associated complications and limitations. The properties of TE scaffolds do not always match the surrounding bone, often sacrificing porosity for improved compressive strength. Previously, the layer-by-layer (LbL) assembly technique was used to deposit nanoclay containing multilayers capable of improving the mechanical properties of open-cell structures without greatly affecting the porosity. However, the previous coatings studied contained poly(ethylenimine) (PEI), which is known to be cytotoxic due to the presence of amine groups, rendering it unsuitable for use in biomedical applications. In this work, poly(diallydimethylammonium chloride) (PDDA)- and chitosan (CHI)-based polyelectrolyte systems were investigated for the purpose of nanoclay addition as an alternative to PEI-based polyelectrolyte systems. Nanocomposite coatings comprising of PEI, poly(acrylic acid) (PAA), Na+ montmorillonite (NC), PDDA, CHI and sodium alginate (ALG) were fabricated. The coatings were deposited in the following manner: (PEI/PAA/PEI/NC), PEI-(PDDA/PAA/PDDA/NC) and (CHI/ALG/CHI/ALG). Results from scanning electron microscopy (SEM) and energy-dispersive X-ray (EDX) analyses demonstrated that the nanoclay was successfully incorporated into each polymer bilayer system, creating a nanocomposite coating. Each coating was successful at tailoring the elastic modulus of the open-cell structures, with polyurethane foams exhibiting an increase from 0.15 ± 0.10 MPa when uncoated to 5.51 ± 0.40 MPa, 6.01 ± 0.36 MPa and 2.61 ± 0.41 MPa when coated with (PEI/PAA/PEI/NC), PEI-(PDDA/PAA/PDDA/NC) and (CHI/ALG/CHI/ALG), respectively. Several biological studies were conducted to determine the cytotoxicity of the coatings, including a resazurin reduction assay, scanning electron microscopy and fluorescent staining of the cell-seeded substrates. In this work, the PDDA-based system exhibited equivalent physical and mechanical properties to the PEI-based system and was significantly more biocompatible, making it a much more suitable alternative for biomaterial applications. Full article
(This article belongs to the Special Issue Multifunctional Scaffolds for Musculoskeletal Regeneration)
Show Figures

Graphical abstract

16 pages, 3275 KiB  
Article
Effects of Different Titanium Surfaces Created by 3D Printing Methods, Particle Sizes, and Acid Etching on Protein Adsorption and Cell Adhesion, Proliferation, and Differentiation
by Max Jin, Haseung Chung, Patrick Kwon and Adil Akkouch
Bioengineering 2022, 9(10), 514; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering9100514 - 28 Sep 2022
Cited by 4 | Viewed by 1585
Abstract
The surfaces of 3D printed titanium prostheses have major impacts on the clinical performance of the prostheses. To investigate the surface effects of the products generated by 3D printed titanium on osseointegration, six surface types of titanium discs produced by the direct metal [...] Read more.
The surfaces of 3D printed titanium prostheses have major impacts on the clinical performance of the prostheses. To investigate the surface effects of the products generated by 3D printed titanium on osseointegration, six surface types of titanium discs produced by the direct metal laser sintering (DMLS) and electron beam melting (EBM) methods, with two sizes of titanium particles and post-printing acid etching, were used to examine the surface topography and to explore the protein adsorption, pro-inflammatory cytokine gene expressions, and MC3T3-E1 cell adhesion, proliferation, and differentiation. The EBM-printed disc showed a stripy and smooth surface without evidence of the particles used, while the DMLS surface contained many particles. After acid etching, small particles on the DMLS surface were removed, whereas the large particles were left. Moreover, distinct proteins with low molecular weights were attached to the 3D printed titanium discs but not to the pre-printing titanium particles. The small titanium particles stimulated the highest TNF-α and IL-6 gene expressions at 24 h. The alizarin red content and osteocalcin gene expression at day 21 were the highest in the groups of acid-etched discs printed by DMLS with the small particles and by EBM. Therefore, the acid-treated surfaces without particles favor osteogenic differentiation. The surface design of 3D printed titanium prostheses should be based on their clinical applications. Full article
(This article belongs to the Special Issue Multifunctional Scaffolds for Musculoskeletal Regeneration)
Show Figures

Graphical abstract

16 pages, 3315 KiB  
Article
Human Adipose-Derived Stromal Cells Delivered on Decellularized Muscle Improve Muscle Regeneration and Regulate RAGE and P38 MAPK
by Lucas C. Olson, James T. Redden, LaStar Gilliam, Tri M. Nguyen, Josephina A. Vossen, David J. Cohen, Zvi Schwartz and Michael J. McClure
Bioengineering 2022, 9(9), 426; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering9090426 - 30 Aug 2022
Cited by 2 | Viewed by 1712
Abstract
Volumetric muscle loss (VML) is the acute loss of muscle mass due to trauma. Such injuries occur primarily in the extremities and are debilitating, as there is no clinical treatment to restore muscle function. Pro-inflammatory advanced glycation end-products (AGEs) and the soluble receptor [...] Read more.
Volumetric muscle loss (VML) is the acute loss of muscle mass due to trauma. Such injuries occur primarily in the extremities and are debilitating, as there is no clinical treatment to restore muscle function. Pro-inflammatory advanced glycation end-products (AGEs) and the soluble receptor for advanced glycation end-products (RAGE) are known to increase in acute trauma patient’s serum and are correlated with increased injury severity. However, it is unclear whether AGEs and RAGE increase in muscle post-trauma. To test this, we used decellularized muscle matrix (DMM), a pro-myogenic, non-immunogenic extracellular matrix biomaterial derived from skeletal muscle. We delivered adipose-derived stromal cells (ASCs) and primary myoblasts to support myogenesis and immunomodulation (N = 8 rats/group). DMM non-seeded and seeded grafts were compared to empty defect and sham controls. Then, 56 days after surgery muscle force was assessed, histology characterized, and protein levels for AGEs, RAGE, p38 MAPK, and myosin heavy chains were measured. Overall, our data showed improved muscle regeneration in ASC-treated injury sites and a regulation of RAGE and p38 MAPK signaling, while myoblast-treated injuries resulted in minor improvements. Taken together, these results suggested that ASCs combined with DMM provides a pro-myogenic microenvironment with immunomodulatory capabilities and indicates further exploration of RAGE signaling in VML. Full article
(This article belongs to the Special Issue Multifunctional Scaffolds for Musculoskeletal Regeneration)
Show Figures

Figure 1

20 pages, 4771 KiB  
Article
The Effect of Mesenchymal Stem Cells, Adipose Tissue Derived Stem Cells, and Cellular Stromal Vascular Fraction on the Repair of Acute Anal Sphincter Injury in Rats
by Wenbin Chen, Zijian He, Shuyu Li, Zixin Wu, Jin Tan, Weifeng Yang, Guanwei Li, Xiaoting Pan, Yuying Liu, Feng-Juan Lyu and Wanglin Li
Bioengineering 2022, 9(7), 318; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering9070318 - 15 Jul 2022
Cited by 5 | Viewed by 1988
Abstract
Background: Anal sphincter incontinence (ASI) can cause a serious decline in the quality of life and can cause a socioeconomic burden. Studies have shown that bone marrow mesenchymal stem cells (MSC) have significant therapeutic effects on ASI, but the cost and risk of [...] Read more.
Background: Anal sphincter incontinence (ASI) can cause a serious decline in the quality of life and can cause a socioeconomic burden. Studies have shown that bone marrow mesenchymal stem cells (MSC) have significant therapeutic effects on ASI, but the cost and risk of MSC harvest limit their further application. In contrast, adipose tissue derived stem cells (ADSC) and cellular stromal vascular fraction (CSVF) as stem cell sources have multipotency and the advantage of easy harvest. Objective: Here we aim to investigate the effects of ADSC and CSVF on treating ASI and compare them to that of bone marrow MSC. Methods: Bone marrow MSC, ADSC, and CSVF were obtained and labeled with green fluorescent protein (GFP), and CSVF was labeled with DIL. Sprague Dawley (SD) rats were divided into 5 groups. Four groups were injected with 0.2 mL phosphate buffer saline (PBS), 1 × 107/0.2 mL of MSC, ADSC, or CSVF, respectively, after model establishment. The control group received no treatment. The repair was assessed by anal functional tests and immunostaining on day 5 and day 10 after injection. Results: MSC, ADSC, and CSVF significantly promoted tissue repair and the recovery of muscle contraction and electromyographic activity in ASI. The generation of myosatellite cells by injected MSC, ADSC, and CSVF was found in the wounded area. On day 5, CSVF showed highest therapeutic effect, while on day 10, MSC and ADSC showed higher therapeutic effects than CSVF. When comparing the effects of MSC and ADSC, ADSC was slightly better than MSC in the indexes of anal pressure, etc. Conclusion: ADSC and CVSF are alternative stem cell sources for ASI repair. Full article
(This article belongs to the Special Issue Multifunctional Scaffolds for Musculoskeletal Regeneration)
Show Figures

Figure 1

18 pages, 3121 KiB  
Article
Impact of Electrospun Piezoelectric Core–Shell PVDFhfp/PDMS Mesh on Tenogenic and Inflammatory Gene Expression in Human Adipose-Derived Stem Cells: Comparison of Static Cultivation with Uniaxial Cyclic Tensile Stretching
by Walter Baumgartner, Petra Wolint, Silvan Hofmann, Cléa Nüesch, Maurizio Calcagni, Marzia Brunelli and Johanna Buschmann
Bioengineering 2022, 9(1), 21; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering9010021 - 08 Jan 2022
Cited by 7 | Viewed by 2236
Abstract
Specific microenvironments can trigger stem cell tenogenic differentiation, such as specific substrates or dynamic cell cultivation. Electrospun meshes composed by core–shell fibers (random or aligned; PDMS core; piezoelectric PVDFhfp shell) were fabricated by coaxial electrospinning. Elastic modulus and residual strain were assessed. Human [...] Read more.
Specific microenvironments can trigger stem cell tenogenic differentiation, such as specific substrates or dynamic cell cultivation. Electrospun meshes composed by core–shell fibers (random or aligned; PDMS core; piezoelectric PVDFhfp shell) were fabricated by coaxial electrospinning. Elastic modulus and residual strain were assessed. Human ASCs were seeded on such scaffolds either under static conditions for 1 week or with subsequent 10% dynamic stretching for 10,800 cycles (1 Hz, 3 h), assessing load elongation curves in a Bose® bioreactor system. Gene expression for tenogenic expression, extracellular matrix, remodeling, pro-fibrotic and inflammatory marker genes were assessed (PCR). For cell-seeded meshes, the E modulus increased from 14 ± 3.8 MPa to 31 ± 17 MPa within 3 h, which was not observed for cell-free meshes. Random fibers resulted in higher tenogenic commitment than aligned fibers. Dynamic cultivation significantly enhanced pro-inflammatory markers. Compared to ASCs in culture flasks, ASCs on random meshes under static cultivation showed a significant upregulation of Mohawk, Tenascin-C and Tenomodulin. The tenogenic commitment expressed by human ASCs in contact with random PVDFhfp/PDMS paves the way for using this novel highly elastic material as an implant to be wrapped around a lacerated tendon, envisioned as a functional anti-adhesion membrane. Full article
(This article belongs to the Special Issue Multifunctional Scaffolds for Musculoskeletal Regeneration)
Show Figures

Figure 1

24 pages, 3734 KiB  
Article
Conductive Bioimprint Using Soft Lithography Technique Based on PEDOT:PSS for Biosensing
by Nor Azila Abd. Wahid, Azadeh Hashemi, John J. Evans and Maan M. Alkaisi
Bioengineering 2021, 8(12), 204; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering8120204 - 09 Dec 2021
Cited by 3 | Viewed by 2751
Abstract
Culture platform surface topography plays an important role in the regulation of biological cell behaviour. Understanding the mechanisms behind the roles of surface topography in cell response are central to many developments in a Lab on a Chip, medical implants and biosensors. In [...] Read more.
Culture platform surface topography plays an important role in the regulation of biological cell behaviour. Understanding the mechanisms behind the roles of surface topography in cell response are central to many developments in a Lab on a Chip, medical implants and biosensors. In this work, we report on a novel development of a biocompatible conductive hydrogel (CH) made of poly (3,4-ethylenedioxythiophene):polystyrene sulfonate (PEDOT:PSS) and gelatin with bioimprinted surface features. The bioimprinted CH offers high conductivity, biocompatibility and high replication fidelity suitable for cell culture applications. The bioimprinted conductive hydrogel is developed to investigate biological cells’ response to their morphological footprint and study their growth, adhesion, cell–cell interactions and proliferation as a function of conductivity. Moreover, optimization of the conductive hydrogel mixture plays an important role in achieving high imprinting resolution and conductivity. The reason behind choosing a conducive hydrogel with high resolution surface bioimprints is to improve cell monitoring while mimicking cells’ natural physical environment. Bioimprints which are a 3D replication of cellular morphology have previously been shown to promote cell attachment, proliferation, differentiation and even cell response to drugs. The conductive substrate, on the other hand, enables cell impedance to be measured and monitored, which is indicative of cell viability and spread. Two dimensional profiles of the cross section of a single cell taken via Atomic Force Microscopy (AFM) from the fixed cell on glass, and its replicas on polydimethylsiloxane (PDMS) and conductive hydrogel (CH) show unprecedented replication of cellular features with an average replication fidelity of more than 90%. Furthermore, crosslinking CH films demonstrated a significant increase in electrical conductivity from 10−6 S/cm to 1 S/cm. Conductive bioimprints can provide a suitable platform for biosensing applications and potentially for monitoring implant-tissue reactions in medical devices. Full article
(This article belongs to the Special Issue Multifunctional Scaffolds for Musculoskeletal Regeneration)
Show Figures

Figure 1

14 pages, 3596 KiB  
Article
Biocompatible Customized 3D Bone Scaffolds Treated with CRFP, an Osteogenic Peptide
by Vamiq M. Mustahsan, Amith Anugu, David E. Komatsu, Imin Kao and Srinivas Pentyala
Bioengineering 2021, 8(12), 199; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering8120199 - 30 Nov 2021
Cited by 6 | Viewed by 2785
Abstract
Background: Currently used synthetic bone graft substitutes (BGS) are either too weak to bear the principal load or if metallic, they can support loading, but can lead to stress shielding and are unable to integrate fully. In this study, we developed biocompatible, 3D [...] Read more.
Background: Currently used synthetic bone graft substitutes (BGS) are either too weak to bear the principal load or if metallic, they can support loading, but can lead to stress shielding and are unable to integrate fully. In this study, we developed biocompatible, 3D printed scaffolds derived from µCT images of the bone that can overcome these issues and support the growth of osteoblasts. Methods: Cylindrical scaffolds were fabricated with acrylonitrile butadiene styrene (ABS) and Stratasys® MED 610 (MED610) materials. The 3D-printed scaffolds were seeded with Mus musculus calvaria cells (MC3T3). After the cells attained confluence, osteogenesis was induced with and without the addition of calcitonin receptor fragment peptide (CRFP) and the bone matrix production was analyzed. Mechanical compression testing was carried out to measure compressive strength, stiffness, and elastic modulus. Results: For the ABS scaffolds, there was a 9.8% increase in compressive strength (p < 0.05) in the scaffolds with no pre-coating and the treatment with CRFP, compared to non-treated scaffolds. Similarly, MED610 scaffolds treated with CRFP showed an 11.9% (polylysine pre-coating) and a 20% (no pre-coating) increase (p < 0.01) in compressive strength compared to non-treated scaffolds. Conclusions: MED610 scaffolds are excellent BGS as they support osteoblast growth and show enhanced bone growth with enhanced compressive strength when augmented with CRFP. Full article
(This article belongs to the Special Issue Multifunctional Scaffolds for Musculoskeletal Regeneration)
Show Figures

Figure 1

22 pages, 8146 KiB  
Article
Dual Network Composites of Poly(vinyl alcohol)-Calcium Metaphosphate/Alginate with Osteogenic Ions for Bone Tissue Engineering in Oral and Maxillofacial Surgery
by Lilis Iskandar, Lucy DiSilvio, Jonathan Acheson and Sanjukta Deb
Bioengineering 2021, 8(8), 107; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering8080107 - 28 Jul 2021
Cited by 2 | Viewed by 3148
Abstract
Despite considerable advances in biomaterials-based bone tissue engineering technologies, autografts remain the gold standard for rehabilitating critical-sized bone defects in the oral and maxillofacial (OMF) region. A majority of advanced synthetic bone substitutes (SBS’s) have not transcended the pre-clinical stage due to inferior [...] Read more.
Despite considerable advances in biomaterials-based bone tissue engineering technologies, autografts remain the gold standard for rehabilitating critical-sized bone defects in the oral and maxillofacial (OMF) region. A majority of advanced synthetic bone substitutes (SBS’s) have not transcended the pre-clinical stage due to inferior clinical performance and translational barriers, which include low scalability, high cost, regulatory restrictions, limited advanced facilities and human resources. The aim of this study is to develop clinically viable alternatives to address the challenges of bone tissue regeneration in the OMF region by developing ‘dual network composites’ (DNC’s) of calcium metaphosphate (CMP)—poly(vinyl alcohol) (PVA)/alginate with osteogenic ions: calcium, zinc and strontium. To fabricate DNC’s, single network composites of PVA/CMP with 10% (w/v) gelatine particles as porogen were developed using two freeze–thawing cycles and subsequently interpenetrated by guluronate-dominant sodium alginate and chelated with calcium, zinc or strontium ions. Physicochemical, compressive, water uptake, thermal, morphological and in vitro biological properties of DNC’s were characterised. The results demonstrated elastic 3D porous scaffolds resembling a ‘spongy bone’ with fluid absorbing capacity, easily sculptable to fit anatomically complex bone defects, biocompatible and osteoconductive in vitro, thus yielding potentially clinically viable for SBS alternatives in OMF surgery. Full article
(This article belongs to the Special Issue Multifunctional Scaffolds for Musculoskeletal Regeneration)
Show Figures

Figure 1

Review

Jump to: Research

22 pages, 1040 KiB  
Review
Tissue Engineering Applied to Skeletal Muscle: Strategies and Perspectives
by Ana Luisa Lopes Martins, Luciana Pastena Giorno and Arnaldo Rodrigues Santos, Jr.
Bioengineering 2022, 9(12), 744; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering9120744 - 30 Nov 2022
Cited by 3 | Viewed by 2465
Abstract
Muscle tissue is formed by elongated and contractile cells with specific morphofunctional characteristics. Thus, it is divided into three basic types: smooth muscle tissue, cardiac striated muscle tissue and skeletal striated muscle tissue. The striated skeletal muscle tissue presents high plasticity, regeneration and [...] Read more.
Muscle tissue is formed by elongated and contractile cells with specific morphofunctional characteristics. Thus, it is divided into three basic types: smooth muscle tissue, cardiac striated muscle tissue and skeletal striated muscle tissue. The striated skeletal muscle tissue presents high plasticity, regeneration and growth capacity due to the presence of satellite cells, quiescent myoblasts that are activated in case of injury to the tissue and originate new muscle fibers when they differentiate. In more severe deficiencies or injuries there is a loss of their regenerative capacity, thus compromising the body’s functionality at different levels. Tissue engineering studies the development of biomaterials capable of stimulating the recovery of cellular activity in injured body tissues, as well as the activity of cells with muscle differentiation potential in injury repair. However, the need for three-dimensional re-assembly in a complex organization makes it difficult to mimic this tissue and fully regenerate it for the sake of precise and effective movements. Thus, this article aims to provide a narrative review of tissue engineering strategies applied to the regeneration of skeletal muscle, in a critical evaluation of research, whether aimed at injury or atrophies such as spinal muscular atrophy. Full article
(This article belongs to the Special Issue Multifunctional Scaffolds for Musculoskeletal Regeneration)
Show Figures

Graphical abstract

33 pages, 4118 KiB  
Review
3D-Printing for Critical Sized Bone Defects: Current Concepts and Future Directions
by Cory K. Mayfield, Mina Ayad, Elizabeth Lechtholz-Zey, Yong Chen and Jay R. Lieberman
Bioengineering 2022, 9(11), 680; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering9110680 - 11 Nov 2022
Cited by 11 | Viewed by 3043
Abstract
The management and definitive treatment of segmental bone defects in the setting of acute trauma, fracture non-union, revision joint arthroplasty, and tumor surgery are challenging clinical problems with no consistently satisfactory solution. Orthopaedic surgeons are developing novel strategies to treat these problems, including [...] Read more.
The management and definitive treatment of segmental bone defects in the setting of acute trauma, fracture non-union, revision joint arthroplasty, and tumor surgery are challenging clinical problems with no consistently satisfactory solution. Orthopaedic surgeons are developing novel strategies to treat these problems, including three-dimensional (3D) printing combined with growth factors and/or cells. This article reviews the current strategies for management of segmental bone loss in orthopaedic surgery, including graft selection, bone graft substitutes, and operative techniques. Furthermore, we highlight 3D printing as a technology that may serve a major role in the management of segmental defects. The optimization of a 3D-printed scaffold design through printing technique, material selection, and scaffold geometry, as well as biologic additives to enhance bone regeneration and incorporation could change the treatment paradigm for these difficult bone repair problems. Full article
(This article belongs to the Special Issue Multifunctional Scaffolds for Musculoskeletal Regeneration)
Show Figures

Figure 1

25 pages, 5165 KiB  
Review
Gelatin Methacryloyl Hydrogels for Musculoskeletal Tissue Regeneration
by Yang-Hee Kim, Jonathan I. Dawson, Richard O. C. Oreffo, Yasuhiko Tabata, Dhiraj Kumar, Conrado Aparicio and Isha Mutreja
Bioengineering 2022, 9(7), 332; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering9070332 - 21 Jul 2022
Cited by 6 | Viewed by 3686
Abstract
Musculoskeletal disorders are a significant burden on the global economy and public health. Hydrogels have significant potential for enhancing the repair of damaged and injured musculoskeletal tissues as cell or drug delivery systems. Hydrogels have unique physicochemical properties which make them promising platforms [...] Read more.
Musculoskeletal disorders are a significant burden on the global economy and public health. Hydrogels have significant potential for enhancing the repair of damaged and injured musculoskeletal tissues as cell or drug delivery systems. Hydrogels have unique physicochemical properties which make them promising platforms for controlling cell functions. Gelatin methacryloyl (GelMA) hydrogel in particular has been extensively investigated as a promising biomaterial due to its tuneable and beneficial properties and has been widely used in different biomedical applications. In this review, a detailed overview of GelMA synthesis, hydrogel design and applications in regenerative medicine is provided. After summarising recent progress in hydrogels more broadly, we highlight recent advances of GelMA hydrogels in the emerging fields of musculoskeletal drug delivery, involving therapeutic drugs (e.g., growth factors, antimicrobial molecules, immunomodulatory drugs and cells), delivery approaches (e.g., single-, dual-release system), and material design (e.g., addition of organic or inorganic materials, 3D printing). The review concludes with future perspectives and associated challenges for developing local drug delivery for musculoskeletal applications. Full article
(This article belongs to the Special Issue Multifunctional Scaffolds for Musculoskeletal Regeneration)
Show Figures

Graphical abstract

31 pages, 6146 KiB  
Review
Biodegradable and Biocompatible Adhesives for the Effective Stabilisation, Repair and Regeneration of Bone
by Antzela Tzagiollari, Helen O. McCarthy, Tanya J. Levingstone and Nicholas J. Dunne
Bioengineering 2022, 9(6), 250; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering9060250 - 10 Jun 2022
Cited by 13 | Viewed by 4002
Abstract
Bone defects and complex fractures present significant challenges for orthopaedic surgeons. Current surgical procedures involve the reconstruction and mechanical stabilisation of complex fractures using metal hardware (i.e., wires, plates and screws). However, these procedures often result in poor healing. An injectable, biocompatible, biodegradable [...] Read more.
Bone defects and complex fractures present significant challenges for orthopaedic surgeons. Current surgical procedures involve the reconstruction and mechanical stabilisation of complex fractures using metal hardware (i.e., wires, plates and screws). However, these procedures often result in poor healing. An injectable, biocompatible, biodegradable bone adhesive that could glue bone fragments back together would present a highly attractive solution. A bone adhesive that meets the many clinical requirements for such an application has yet to be developed. While synthetic and biological polymer-based adhesives (e.g., cyanoacrylates, PMMA, fibrin, etc.) have been used effectively as bone void fillers, these materials lack biomechanical integrity and demonstrate poor injectability, which limits the clinical effectiveness and potential for minimally invasive delivery. This systematic review summarises conventional approaches and recent developments in the area of bone adhesives for orthopaedic applications. The required properties for successful bone repair adhesives, which include suitable injectability, setting characteristics, mechanical properties, biocompatibility and an ability to promote new bone formation, are highlighted. Finally, the potential to achieve repair of challenging bone voids and fractures as well as the potential of new bioinspired adhesives and the future directions relating to their clinical development are discussed. Full article
(This article belongs to the Special Issue Multifunctional Scaffolds for Musculoskeletal Regeneration)
Show Figures

Figure 1

20 pages, 2321 KiB  
Review
Biodegradable Magnesium Biomaterials—Road to the Clinic
by Shukufe Amukarimi and Masoud Mozafari
Bioengineering 2022, 9(3), 107; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering9030107 - 05 Mar 2022
Cited by 32 | Viewed by 4476
Abstract
In recent decades, we have witnessed radical changes in the use of permanent biomaterials. The intrinsic ability of magnesium (Mg) and its alloys to degrade without releasing toxic degradation products has led to a vast range of applications in the biomedical field, including [...] Read more.
In recent decades, we have witnessed radical changes in the use of permanent biomaterials. The intrinsic ability of magnesium (Mg) and its alloys to degrade without releasing toxic degradation products has led to a vast range of applications in the biomedical field, including cardiovascular stents, musculoskeletal, and orthopedic applications. With the use of biodegradable Mg biomaterials, patients would not suffer second surgery and surgical pain anymore. Be that as it may, the main drawbacks of these biomaterials are the high corrosion rate and unexpected degradation in physiological environments. Since biodegradable Mg-based implants are expected to show controllable degradation and match the requirements of specific applications, various techniques, such as designing a magnesium alloy and modifying the surface characteristics, are employed to tailor the degradation rate. In this paper, some fundamentals and particular aspects of magnesium degradation in physiological environments are summarized, and approaches to control the degradation behavior of Mg-based biomaterials are presented. Full article
(This article belongs to the Special Issue Multifunctional Scaffolds for Musculoskeletal Regeneration)
Show Figures

Figure 1

20 pages, 1129 KiB  
Review
Lumbar Intervertebral Disc Herniation: Annular Closure Devices and Key Design Requirements
by Alexandra Alcántara Guardado, Alexander Baker, Andrew Weightman, Judith A. Hoyland and Glen Cooper
Bioengineering 2022, 9(2), 47; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering9020047 - 19 Jan 2022
Cited by 7 | Viewed by 7628
Abstract
Lumbar disc herniation is one of the most common degenerative spinal conditions resulting in lower back pain and sciatica. Surgical treatment options include microdiscectomy, lumbar fusion, total disc replacement, and other minimally invasive approaches. At present, microdiscectomy procedures are the most used technique; [...] Read more.
Lumbar disc herniation is one of the most common degenerative spinal conditions resulting in lower back pain and sciatica. Surgical treatment options include microdiscectomy, lumbar fusion, total disc replacement, and other minimally invasive approaches. At present, microdiscectomy procedures are the most used technique; however, the annulus fibrosus is left with a defect that without treatment may contribute to high reherniation rates and changes in the biomechanics of the lumbar spine. This paper aims to review current commercially available products that mechanically close the annulus including the AnchorKnot® suture-passing device and the Barricaid® annular closure device. Previous studies and reviews have focused mainly on a biomimetic biomaterials approach and have described some mechanical and biological requirements for an active annular repair/regeneration strategy but are still far away from clinical implementation. Therefore, in this paper we aim to create a design specification for a mechanical annular closure strategy by identifying the most important mechanical and biological design parameters, including consideration of material selection, preclinical testing requirements, and requirements for clinical implementation. Full article
(This article belongs to the Special Issue Multifunctional Scaffolds for Musculoskeletal Regeneration)
Show Figures

Figure 1

27 pages, 1780 KiB  
Review
Tissue Engineering Strategies for Treating Avascular Necrosis of the Femoral Head
by Sumit Murab, Teresa Hawk, Alexander Snyder, Sydney Herold, Meghana Totapally and Patrick W. Whitlock
Bioengineering 2021, 8(12), 200; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering8120200 - 02 Dec 2021
Cited by 9 | Viewed by 9649
Abstract
Avascular necrosis (AVN) of the femoral head commonly leads to symptomatic osteoarthritis of the hip. In older patients, hip replacement is a viable option that restores the hip biomechanics and improves pain but in pediatric, adolescent, and young adult patients hip replacements impose [...] Read more.
Avascular necrosis (AVN) of the femoral head commonly leads to symptomatic osteoarthritis of the hip. In older patients, hip replacement is a viable option that restores the hip biomechanics and improves pain but in pediatric, adolescent, and young adult patients hip replacements impose significant activity limitations and the need for multiple revision surgeries with increasing risk of complication. Early detection of AVN requires a high level of suspicion as diagnostic techniques such as X-rays are not sensitive in the early stages of the disease. There are multiple etiologies that can lead to this disease. In the pediatric and adolescent population, trauma is a commonly recognized cause of AVN. The understanding of the pathophysiology of the disease is limited, adding to the challenge of devising a clinically effective treatment strategy. Surgical techniques to prevent progression of the disease and avoid total hip replacement include core decompression, vascular grafts, and use of bone-marrow derived stem cells with or without adjuncts, such as bisphosphonates and bone morphogenetic protein (BMP), all of which are partially effective only in the very early stages of the disease. Further, these strategies often only improve pain and range of motion in the short-term in some patients and do not predictably prevent progression of the disease. Tissue engineering strategies with the combined use of biomaterials, stem cells and growth factors offer a potential strategy to avoid metallic implants and surgery. Structural, bioactive biomaterial platforms could help in stabilizing the femoral head while inducing osteogenic differentiation to regenerate bone and provide angiogenic cues to concomitantly recover vasculature in the femoral head. Moreover, injectable systems that can be delivered using a minimal invasive procedure and provide mechanical support the collapsing femoral head could potentially alleviate the need for surgical interventions in the future. The present review describes the limitations of existing surgical methods and the recent advances in tissue engineering that are leading in the direction of a clinically effective, translational solution for AVN in future. Full article
(This article belongs to the Special Issue Multifunctional Scaffolds for Musculoskeletal Regeneration)
Show Figures

Figure 1

28 pages, 2900 KiB  
Review
Bioactive Scaffolds Integrated with Liposomal or Extracellular Vesicles for Bone Regeneration
by Minjee Kang, Chung-Sung Lee and Min Lee
Bioengineering 2021, 8(10), 137; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering8100137 - 01 Oct 2021
Cited by 31 | Viewed by 6234
Abstract
With population aging and increased life expectancy, an increasing number of people are facing musculoskeletal health problems that necessitate therapeutic intervention at defect sites. Bone tissue engineering (BTE) has become a promising approach for bone graft substitutes as traditional treatments using autografts or [...] Read more.
With population aging and increased life expectancy, an increasing number of people are facing musculoskeletal health problems that necessitate therapeutic intervention at defect sites. Bone tissue engineering (BTE) has become a promising approach for bone graft substitutes as traditional treatments using autografts or allografts involve clinical complications. Significant advancements have been made in developing ideal BTE scaffolds that can integrate bioactive molecules promoting robust bone repair. Herein, we review bioactive scaffolds tuned for local bone regenerative therapy, particularly through integrating synthetic liposomal vesicles or extracellular vesicles to the scaffolds. Liposomes offer an excellent drug delivery system providing sustained release of the loaded bioactive molecules. Extracellular vesicles, with their inherent capacity to carry bioactive molecules, are emerging as an advanced substitute of synthetic nanoparticles and a novel cell-free therapy for bone regeneration. We discuss the recent advance in the use of synthetic liposomes and extracellular vesicles as bioactive materials combined with scaffolds, highlighting major challenges and opportunities for their applications in bone regeneration. We put a particular focus on strategies to integrate vesicles to various biomaterial scaffolds and introduce the latest advances in achieving sustained release of bioactive molecules from the vesicle-loaded scaffolds at the bone defect site. Full article
(This article belongs to the Special Issue Multifunctional Scaffolds for Musculoskeletal Regeneration)
Show Figures

Figure 1

Back to TopTop