ijms-logo

Journal Browser

Journal Browser

Novel Biomaterials for Tissue Engineering 2018

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Materials Science".

Deadline for manuscript submissions: closed (25 April 2018) | Viewed by 183161

Printed Edition Available!
A printed edition of this Special Issue is available here.

Special Issue Editor

Special Issue Information

Dear Colleagues,

This Special Issue, “Novel Biomaterials for Tissue Engineering”, will cover a selection of recent research topics and current review articles in the field biomaterials and for tissue engineering and regeneration purposes. Experimental papers, up-to-date review articles, and commentaries are all welcome.

The concept of regenerating tissues, with properties and functions that mimic natural tissues, has attracted significant attention in recent years. It provides potential solutions for many diseases treatment and other healthcare problems. To fully realize the potential of the approach, it is crucial to have a rational biomaterial design to create novel scaffolds, and other materials systems suitable for tissue engineering, repair and regeneration. Research advances on the topic include the design of new biomaterials and their composites, the scaffold fabrication via subtractive and additive manufacturing approaches, the development of implantable scaffolds for disease monitoring, diagnostics, and treatment, as well as the understanding of cells-biomaterial scaffolds interaction.

Dr. Emmanuel Stratakis
Guest Editor

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. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. 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

  • biomaterials

  • tissue engineering

  • tissue regeneration

  • scaffolds

  • bioprinting

  • biomaterial structuring

  • cell-biomaterial interaction

  • implantable scaffolds

Related Special Issue

Published Papers (26 papers)

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

Editorial

Jump to: Research, Review, Other

4 pages, 179 KiB  
Editorial
Novel Biomaterials for Tissue Engineering 2018
by Emmanuel Stratakis
Int. J. Mol. Sci. 2018, 19(12), 3960; https://doi.org/10.3390/ijms19123960 - 09 Dec 2018
Cited by 19 | Viewed by 3072
Abstract
The concept of regenerating tissues, with properties and functions that mimic natural tissues, has attracted significant attention in recent years. [...] Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)

Research

Jump to: Editorial, Review, Other

21 pages, 6844 KiB  
Article
Engineering Cell Adhesion and Orientation via Ultrafast Laser Fabricated Microstructured Substrates
by Eleftheria Babaliari, Paraskevi Kavatzikidou, Despoina Angelaki, Lefki Chaniotaki, Alexandra Manousaki, Alexandra Siakouli-Galanopoulou, Anthi Ranella and Emmanuel Stratakis
Int. J. Mol. Sci. 2018, 19(7), 2053; https://doi.org/10.3390/ijms19072053 - 14 Jul 2018
Cited by 25 | Viewed by 5279
Abstract
Cell responses depend on the stimuli received by the surrounding extracellular environment, which provides the cues required for adhesion, orientation, proliferation, and differentiation at the micro and the nano scales. In this study, discontinuous microcones on silicon (Si) and continuous microgrooves on polyethylene [...] Read more.
Cell responses depend on the stimuli received by the surrounding extracellular environment, which provides the cues required for adhesion, orientation, proliferation, and differentiation at the micro and the nano scales. In this study, discontinuous microcones on silicon (Si) and continuous microgrooves on polyethylene terephthalate (PET) substrates were fabricated via ultrashort pulsed laser irradiation at various fluences, resulting in microstructures with different magnitudes of roughness and varying geometrical characteristics. The topographical models attained were specifically developed to imitate the guidance and alignment of Schwann cells for the oriented axonal regrowth that occurs in nerve regeneration. At the same time, positive replicas of the silicon microstructures were successfully reproduced via soft lithography on the biodegradable polymer poly(lactide-co-glycolide) (PLGA). The anisotropic continuous (PET) and discontinuous (PLGA replicas) microstructured polymeric substrates were assessed in terms of their influence on Schwann cell responses. It is shown that the micropatterned substrates enable control over cellular adhesion, proliferation, and orientation, and are thus useful to engineer cell alignment in vitro. This property is potentially useful in the fields of neural tissue engineering and for dynamic microenvironment systems that simulate in vivo conditions. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Graphical abstract

16 pages, 2289 KiB  
Article
Application of Millifluidics to Encapsulate and Support Viable Human Mesenchymal Stem Cells in a Polysaccharide Hydrogel
by Fabien Nativel, Denis Renard, Fahd Hached, Pierre-Gabriel Pinta, Cyril D’Arros, Pierre Weiss, Catherine Le Visage, Jérôme Guicheux, Aurélie Billon-Chabaud and Gael Grimandi
Int. J. Mol. Sci. 2018, 19(7), 1952; https://doi.org/10.3390/ijms19071952 - 03 Jul 2018
Cited by 11 | Viewed by 4878
Abstract
Human adipose-derived stromal cells (hASCs) are widely known for their immunomodulatory and anti-inflammatory properties. This study proposes a method to protect cells during and after their injection by encapsulation in a hydrogel using a droplet millifluidics technique. A biocompatible, self-hardening biomaterial composed of [...] Read more.
Human adipose-derived stromal cells (hASCs) are widely known for their immunomodulatory and anti-inflammatory properties. This study proposes a method to protect cells during and after their injection by encapsulation in a hydrogel using a droplet millifluidics technique. A biocompatible, self-hardening biomaterial composed of silanized-hydroxypropylmethylcellulose (Si-HPMC) hydrogel was used and dispersed in an oil continuous phase. Spherical particles with a mean diameter of 200 μm could be obtained in a reproducible manner. The viability of the encapsulated hASCs in the Si-HPMC particles was 70% after 14 days in vitro, confirming that the Si-HPMC particles supported the diffusion of nutrients, vitamins, and glucose essential for survival of the encapsulated hASCs. The combination of droplet millifluidics and biomaterials is therefore a very promising method for the development of new cellular microenvironments, with the potential for applications in biomedical engineering. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Graphical abstract

21 pages, 4835 KiB  
Article
Collagen as Coating Material for 45S5 Bioactive Glass-Based Scaffolds for Bone Tissue Engineering
by Jasmin Hum and Aldo R. Boccaccini
Int. J. Mol. Sci. 2018, 19(6), 1807; https://doi.org/10.3390/ijms19061807 - 19 Jun 2018
Cited by 47 | Viewed by 7648
Abstract
Highly porous 45S5 bioactive glass-based scaffolds were fabricated by the foam replica technique and coated with collagen by a novel method. After an initial cleaning step of the bioactive glass surface to expose reactive –OH groups, samples were surface functionalized by (3-aminopropyl)triethoxysilane (APTS). [...] Read more.
Highly porous 45S5 bioactive glass-based scaffolds were fabricated by the foam replica technique and coated with collagen by a novel method. After an initial cleaning step of the bioactive glass surface to expose reactive –OH groups, samples were surface functionalized by (3-aminopropyl)triethoxysilane (APTS). Functionalized scaffolds were immersed in a collagen solution, left for gelling at 37 °C, and dried at room temperature. The collagen coating was further stabilized by crosslinking with 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC) and N-hydroxysuccinimide (NHS). Applying this coating method, a layer thickness of a few micrometers was obtained without affecting the overall scaffold macroporosity. In addition, values of compressive strength were enhanced by a factor of five, increasing from 0.04 ± 0.02 MPa for uncoated scaffolds to 0.18 ± 0.03 MPa for crosslinked collagen-coated scaffolds. The composite material developed in this study exhibited positive cell (MG-63) viability as well as suitable cell attachment and proliferation on the surface. The combination of bioactivity, mechanical competence, and cellular response makes this novel scaffold system attractive for bone tissue engineering. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Figure 1

14 pages, 11075 KiB  
Article
3D Bioprinted Artificial Trachea with Epithelial Cells and Chondrogenic-Differentiated Bone Marrow-Derived Mesenchymal Stem Cells
by Sang-Woo Bae, Kang-Woog Lee, Jae-Hyun Park, JunHee Lee, Cho-Rok Jung, JunJie Yu, Hwi-Yool Kim and Dae-Hyun Kim
Int. J. Mol. Sci. 2018, 19(6), 1624; https://doi.org/10.3390/ijms19061624 - 31 May 2018
Cited by 75 | Viewed by 8467
Abstract
Tracheal resection has limited applicability. Although various tracheal replacement strategies were performed using artificial prosthesis, synthetic stents and tissue transplantation, the best method in tracheal reconstruction remains to be identified. Recent advances in tissue engineering enabled 3D bioprinting using various biocompatible materials including [...] Read more.
Tracheal resection has limited applicability. Although various tracheal replacement strategies were performed using artificial prosthesis, synthetic stents and tissue transplantation, the best method in tracheal reconstruction remains to be identified. Recent advances in tissue engineering enabled 3D bioprinting using various biocompatible materials including living cells, thereby making the product clinically applicable. Moreover, clinical interest in mesenchymal stem cell has dramatically increased. Here, rabbit bone marrow-derived mesenchymal stem cells (bMSC) and rabbit respiratory epithelial cells were cultured. The chondrogenic differentiation level of bMSC cultured in regular media (MSC) and that in chondrogenic media (d-MSC) were compared. Dual cell-containing artificial trachea were manufactured using a 3D bioprinting method with epithelial cells and undifferentiated bMSC (MSC group, n = 6) or with epithelial cells and chondrogenic-differentiated bMSC (d-MSC group, n = 6). d-MSC showed a relatively higher level of glycosaminoglycan (GAG) accumulation and chondrogenic marker gene expression than MSC in vitro. Neo-epithelialization and neo-vascularization were observed in all groups in vivo but neo-cartilage formation was only noted in d-MSC. The epithelial cells in the 3D bioprinted artificial trachea were effective in respiratory epithelium regeneration. Chondrogenic-differentiated bMSC had more neo-cartilage formation potential in a short period. Nevertheless, the cartilage formation was observed only in a localized area. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Figure 1

14 pages, 3210 KiB  
Article
Healing of Osteochondral Defects Implanted with Biomimetic Scaffolds of Poly(ε-Caprolactone)/Hydroxyapatite and Glycidyl-Methacrylate-Modified Hyaluronic Acid in a Minipig
by Yi-Ho Hsieh, Bo-Yuan Shen, Yao-Horng Wang, Bojain Lin, Hung-Maan Lee and Ming-Fa Hsieh
Int. J. Mol. Sci. 2018, 19(4), 1125; https://doi.org/10.3390/ijms19041125 - 09 Apr 2018
Cited by 40 | Viewed by 5437
Abstract
Articular cartilage is a structure lack of vascular distribution. Once the cartilage is injured or diseased, it is unable to regenerate by itself. Surgical treatments do not effectively heal defects in articular cartilage. Tissue engineering is the most potential solution to this problem. [...] Read more.
Articular cartilage is a structure lack of vascular distribution. Once the cartilage is injured or diseased, it is unable to regenerate by itself. Surgical treatments do not effectively heal defects in articular cartilage. Tissue engineering is the most potential solution to this problem. In this study, methoxy poly(ethylene glycol)-block-poly(ε-caprolactone) (mPEG-PCL) and hydroxyapatite at a weight ratio of 2:1 were mixed via fused deposition modeling (FDM) layer by layer to form a solid scaffold. The scaffolds were further infiltrated with glycidyl methacrylate hyaluronic acid loading with 10 ng/mL of Transforming Growth Factor-β1 and photo cross-linked on top of the scaffolds. An in vivo test was performed on the knees of Lanyu miniature pigs for a period of 12 months. The healing process of the osteochondral defects was followed by computer tomography (CT). The defect was fully covered with regenerated tissues in the control pig, while different tissues were grown in the defect of knee of the experimental pig. In the gross anatomy of the cross section, the scaffold remained in the subchondral location, while surface cartilage was regenerated. The cross section of the knees of both the control and experimental pigs were subjected to hematoxylin and eosin staining. The cartilage of the knee in the experimental pig was partially matured, e.g., few chondrocyte cells were enclosed in the lacunae. In the knee of the control pig, the defect was fully grown with fibrocartilage. In another in vivo experiment in a rabbit and a pig, the composite of the TGF-β1-loaded hydrogel and scaffolds was found to regenerate hyaline cartilage. However, scaffolds that remain in the subchondral lesion potentially delay the healing process. Therefore, the structural design of the scaffold should be reconsidered to match the regeneration process of both cartilage and subchondral bone. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Graphical abstract

14 pages, 6229 KiB  
Article
Electrophoretic Deposition of Hydroxyapatite Film Containing Re-Doped MoS2 Nanoparticles
by Hila Shalom, Yishay Feldman, Rita Rosentsveig, Iddo Pinkas, Ifat Kaplan-Ashiri, Alexey Moshkovich, Vladislav Perfilyev, Lev Rapoport and Reshef Tenne
Int. J. Mol. Sci. 2018, 19(3), 657; https://doi.org/10.3390/ijms19030657 - 26 Feb 2018
Cited by 13 | Viewed by 4590
Abstract
Films combining hydroxyapatite (HA) with minute amounts (ca. 1 weight %) of (rhenium doped) fullerene-like MoS2 (IF) nanoparticles were deposited onto porous titanium substrate through electrophoretic process (EPD). The films were analyzed by scanning electron microscopy (SEM), X-ray diffraction and Raman spectroscopy. [...] Read more.
Films combining hydroxyapatite (HA) with minute amounts (ca. 1 weight %) of (rhenium doped) fullerene-like MoS2 (IF) nanoparticles were deposited onto porous titanium substrate through electrophoretic process (EPD). The films were analyzed by scanning electron microscopy (SEM), X-ray diffraction and Raman spectroscopy. The SEM analysis showed relatively uniform coatings of the HA + IF on the titanium substrate. Chemical composition analysis using energy dispersive X-ray spectroscopy (EDS) of the coatings revealed the presence of calcium phosphate minerals like hydroxyapatite, as a majority phase. Tribological tests were undertaken showing that the IF nanoparticles endow the HA film very low friction and wear characteristics. Such films could be of interest for various medical technologies. Means for improving the adhesion of the film to the underlying substrate and its fracture toughness, without compromising its biocompatibility are discussed at the end. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Figure 1

18 pages, 11300 KiB  
Article
3D Biomimetic Magnetic Structures for Static Magnetic Field Stimulation of Osteogenesis
by Irina Alexandra Paun, Roxana Cristina Popescu, Bogdan Stefanita Calin, Cosmin Catalin Mustaciosu, Maria Dinescu and Catalin Romeo Luculescu
Int. J. Mol. Sci. 2018, 19(2), 495; https://doi.org/10.3390/ijms19020495 - 07 Feb 2018
Cited by 37 | Viewed by 6147
Abstract
We designed, fabricated and optimized 3D biomimetic magnetic structures that stimulate the osteogenesis in static magnetic fields. The structures were fabricated by direct laser writing via two-photon polymerization of IP-L780 photopolymer and were based on ellipsoidal, hexagonal units organized in a multilayered architecture. [...] Read more.
We designed, fabricated and optimized 3D biomimetic magnetic structures that stimulate the osteogenesis in static magnetic fields. The structures were fabricated by direct laser writing via two-photon polymerization of IP-L780 photopolymer and were based on ellipsoidal, hexagonal units organized in a multilayered architecture. The magnetic activity of the structures was assured by coating with a thin layer of collagen-chitosan-hydroxyapatite-magnetic nanoparticles composite. In vitro experiments using MG-63 osteoblast-like cells for 3D structures with gradients of pore size helped us to find an optimum pore size between 20–40 µm. Starting from optimized 3D structures, we evaluated both qualitatively and quantitatively the effects of static magnetic fields of up to 250 mT on cell proliferation and differentiation, by ALP (alkaline phosphatase) production, Alizarin Red and osteocalcin secretion measurements. We demonstrated that the synergic effect of 3D structure optimization and static magnetic stimulation enhances the bone regeneration by a factor greater than 2 as compared with the same structure in the absence of a magnetic field. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Graphical abstract

14 pages, 3986 KiB  
Article
Aging Donor-Derived Human Mesenchymal Stem Cells Exhibit Reduced Reactive Oxygen Species Loads and Increased Differentiation Potential Following Serial Expansion on a PEG-PCL Copolymer Substrate
by Daniel A. Balikov, Spencer W. Crowder, Jung Bok Lee, Yunki Lee, Ung Hyun Ko, Mi-Lan Kang, Won Shik Kim, Jennifer H. Shin and Hak-Joon Sung
Int. J. Mol. Sci. 2018, 19(2), 359; https://doi.org/10.3390/ijms19020359 - 25 Jan 2018
Cited by 7 | Viewed by 4688
Abstract
Human mesenchymal stem cells (hMSCs) have been widely studied for therapeutic development in tissue engineering and regenerative medicine. They can be harvested from human donors via tissue biopsies, such as bone marrow aspiration, and cultured to reach clinically relevant cell numbers. However, an [...] Read more.
Human mesenchymal stem cells (hMSCs) have been widely studied for therapeutic development in tissue engineering and regenerative medicine. They can be harvested from human donors via tissue biopsies, such as bone marrow aspiration, and cultured to reach clinically relevant cell numbers. However, an unmet issue lies in the fact that the hMSC donors for regenerative therapies are more likely to be of advanced age. Their stem cells are not as potent compared to those of young donors, and continue to lose healthy, stemness-related activities when the hMSCs are serially passaged in tissue culture plates. Here, we have developed a cheap, scalable, and effective copolymer film to culture hMSCs obtained from aged human donors over several passages without loss of reactive oxygen species (ROS) handling or differentiation capacity. Assays of cell morphology, reactive oxygen species load, and differentiation potential demonstrate the effectiveness of copolymer culture on reduction in senescence-related activities of aging donor-derived hMSCs that could hinder the therapeutic potential of autologous stem cell therapies. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Graphical abstract

10 pages, 2697 KiB  
Article
The Osteogenic Differentiation Effect of the FN Type 10-Peptide Amphiphile on PCL Fiber
by Ye-Rang Yun, Hae-Won Kim and Jun-Hyeog Jang
Int. J. Mol. Sci. 2018, 19(1), 153; https://doi.org/10.3390/ijms19010153 - 04 Jan 2018
Cited by 6 | Viewed by 3590
Abstract
The fibronectin type 10-peptide amphiphile (FNIII10-PA) was previously genetically engineered and showed osteogenic differentiation activity on rat bone marrow stem cells (rBMSCs). In this study, we investigated whether FNIII10-PA demonstrated cellular activity on polycaprolactone (PCL) fibers. FNIII10-PA significantly increased protein production and cell [...] Read more.
The fibronectin type 10-peptide amphiphile (FNIII10-PA) was previously genetically engineered and showed osteogenic differentiation activity on rat bone marrow stem cells (rBMSCs). In this study, we investigated whether FNIII10-PA demonstrated cellular activity on polycaprolactone (PCL) fibers. FNIII10-PA significantly increased protein production and cell adhesion activity on PCL fibers in a dose-dependent manner. In cell proliferation results, there was no effect on cell proliferation activity by FNIII10-PA; however, FNIII10-PA induced the osteogenic differentiation of MC3T3-E1 cells via upregulation of bone sialoprotein (BSP), collagen type I (Col I), osteocalcin (OC), osteopontin (OPN), and runt-related transcription factor 2 (Runx2) mitochondrial RNA (mRNA) levels; it did not increase the alkaline phosphatase (ALP) mRNA level. These results indicate that FNIII10-PA has potential as a new biomaterial for bone tissue engineering applications. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Graphical abstract

11119 KiB  
Article
Effects and Mechanisms of Total Flavonoids from Blumea balsamifera (L.) DC. on Skin Wound in Rats
by Yuxin Pang, Yan Zhang, Luqi Huang, Luofeng Xu, Kai Wang, Dan Wang, Lingliang Guan, Yingbo Zhang, Fulai Yu, Zhenxia Chen and Xiaoli Xie
Int. J. Mol. Sci. 2017, 18(12), 2766; https://doi.org/10.3390/ijms18122766 - 19 Dec 2017
Cited by 38 | Viewed by 6728
Abstract
Chinese herbal medicine (CHM) evolved through thousands of years of practice and was popular not only among the Chinese population, but also most countries in the world. Blumea balsamifera (L.) DC. as a traditional treatment for wound healing in Li Nationality Medicine has [...] Read more.
Chinese herbal medicine (CHM) evolved through thousands of years of practice and was popular not only among the Chinese population, but also most countries in the world. Blumea balsamifera (L.) DC. as a traditional treatment for wound healing in Li Nationality Medicine has a long history of nearly 2000 years. This study was to evaluate the effects of total flavonoids from Blumea balsamifera (L.) DC. on skin excisional wound on the back of Sprague-Dawley rats, reveal its chemical constitution, and postulate its action mechanism. The rats were divided into five groups and the model groups were treated with 30% glycerol, the positive control groups with Jing Wan Hong (JWH) ointment, and three treatment groups with high dose (2.52 g·kg−1), medium dose (1.26 g·kg−1), and low dose (0.63 g·kg−1) of total flavonoids from B. balsamifera. During 10 consecutive days of treatment, the therapeutic effects of rates were evaluated. On day 1, day 3, day 5, day 7, and day 10 after treatment, skin samples were taken from all the rats for further study. Significant increases of granulation tissue, fibroblast, and capillary vessel proliferation were observed at day 7 in the high dose and positive control groups, compared with the model group, with the method of 4% paraformaldehyde for histopathological examination and immunofluorescence staining. To reveal the action mechanisms of total flavonoids on wound healing, the levels of CD68, vascular endothelial growth factor (VEGF), transforming growth factor-β1 (TGF-β1), and hydroxyproline were measured at different days. Results showed that total flavonoids had significant effects on rat skin excisional wound healing compared with controls, especially high dose ones (p < 0.05). Furthermore, the total flavonoid extract was investigated phytochemically, and twenty-seven compounds were identified from the total flavonoid sample by ultra-high-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry/diode array detector (UPLC-Q-TOF-MS/DAD), including 16 flavonoid aglucons, five flavonoid glycosides (main peaks in chromatogram), five chlorogenic acid analogs, and 1 coumarin. Reports show that flavonoid glycoside possesses therapeutic effects of curing wounds by inducing neovascularization, and chlorogenic acid also has anti-inflammatory and wound healing activities; we postulated that all the ingredients in total flavonoids sample maybe exert a synergetic effect on wound curing. Accompanied with detection of four growth factors, the upregulation of these key growth factors may be the mechanism of therapeutic activities of total flavonoids. The present study confirmed undoubtedly that flavonoids were the main active constituents that contribute to excisional wound healing, and suggested its action mechanism of improving expression levels of growth factors at different healing phases. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Figure 1

3754 KiB  
Article
Synthesis and Characterization of Nanofunctionalized Gelatin Methacrylate Hydrogels
by Kamel Rahali, Ghazi Ben Messaoud, Cyril J.F. Kahn, Laura Sanchez-Gonzalez, Mouna Kaci, Franck Cleymand, Solenne Fleutot, Michel Linder, Stéphane Desobry and Elmira Arab-Tehrany
Int. J. Mol. Sci. 2017, 18(12), 2675; https://doi.org/10.3390/ijms18122675 - 10 Dec 2017
Cited by 72 | Viewed by 9019
Abstract
Given the importance of the extracellular medium during tissue formation, it was wise to develop an artificial structure that mimics the extracellular matrix while having improved physico-chemical properties. That is why the choice was focused on gelatin methacryloyl (GelMA), an inexpensive biocompatible hydrogel. [...] Read more.
Given the importance of the extracellular medium during tissue formation, it was wise to develop an artificial structure that mimics the extracellular matrix while having improved physico-chemical properties. That is why the choice was focused on gelatin methacryloyl (GelMA), an inexpensive biocompatible hydrogel. Physicochemical and mechanical properties were improved by the incorporation of nanoparticles developed from two innovative fabrication processes: High shear fluid and low frequencies/high frequencies ultrasounds. Both rapeseed nanoliposomes and nanodroplets were successfully incorporated in the GelMA networks during the photo polymerization process. The impact on polymer microstructure was investigated by Fourier-transform infrared spectroscopy (FTIR), scanning electron microscopy (SEM), and enzymatic degradation investigations. Mechanical stability and viscoelastic tests were conducted to demonstrate the beneficial effect of the functionalization on GelMA hydrogels. Adding nanoparticles to GelMA improved the surface properties (porosity), tuned swelling, and degradability properties. In addition, we observed that nanoemulsion didn’t change significantly the mechanical properties to shear and compression solicitations, whereas nanoliposome addition decreased Young’s modulus under compression solicitations. Thus, these ways of functionalization allow controlling the design of the material by choosing the type of nanoparticle (nanoliposome or nanoemulsion) in function of the application. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Figure 1

10738 KiB  
Article
A New Bone Substitute Developed from 3D-Prints of Polylactide (PLA) Loaded with Collagen I: An In Vitro Study
by Ulrike Ritz, Rebekka Gerke, Hermann Götz, Stefan Stein and Pol Maria Rommens
Int. J. Mol. Sci. 2017, 18(12), 2569; https://doi.org/10.3390/ijms18122569 - 29 Nov 2017
Cited by 47 | Viewed by 7326
Abstract
Although a lot of research has been performed, large segmental bone defects caused by trauma, infection, bone tumors or revision surgeries still represent big challenges for trauma surgeons. New and innovative bone substitutes are needed. Three-dimensional (3D) printing is a novel procedure to [...] Read more.
Although a lot of research has been performed, large segmental bone defects caused by trauma, infection, bone tumors or revision surgeries still represent big challenges for trauma surgeons. New and innovative bone substitutes are needed. Three-dimensional (3D) printing is a novel procedure to create 3D porous scaffolds that can be used for bone tissue engineering. In the present study, solid discs as well as porous cage-like 3D prints made of polylactide (PLA) are coated or filled with collagen, respectively, and tested for biocompatibility and endotoxin contamination. Microscopic analyses as well as proliferation assays were performed using various cell types on PLA discs. Stromal-derived factor (SDF-1) release from cages filled with collagen was analyzed and the effect on endothelial cells tested. This study confirms the biocompatibility of PLA and demonstrates an endotoxin contamination clearly below the FDA (Food and Drug Administration) limit. Cells of various cell types (osteoblasts, osteoblast-like cells, fibroblasts and endothelial cells) grow, spread and proliferate on PLA-printed discs. PLA cages loaded with SDF-1 collagen display a steady SDF-1 release, support cell growth of endothelial cells and induce neo-vessel formation. These results demonstrate the potential for PLA scaffolds printed with an inexpensive desktop printer in medical applications, for example, in bone tissue engineering. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Graphical abstract

2074 KiB  
Article
Human Mesenchymal Stem Cells Growth and Osteogenic Differentiation on Piezoelectric Poly(vinylidene fluoride) Microsphere Substrates
by R. Sobreiro-Almeida, M. N. Tamaño-Machiavello, E. O. Carvalho, L. Cordón, S. Doria, L. Senent, D. M. Correia, C. Ribeiro, S. Lanceros-Méndez, R. Sabater i Serra, J. L. Gomez Ribelles and A. Sempere
Int. J. Mol. Sci. 2017, 18(11), 2391; https://doi.org/10.3390/ijms18112391 - 11 Nov 2017
Cited by 34 | Viewed by 4941
Abstract
The aim of this work was to determine the influence of the biomaterial environment on human mesenchymal stem cell (hMSC) fate when cultured in supports with varying topography. Poly(vinylidene fluoride) (PVDF) culture supports were prepared with structures ranging between 2D and 3D, based [...] Read more.
The aim of this work was to determine the influence of the biomaterial environment on human mesenchymal stem cell (hMSC) fate when cultured in supports with varying topography. Poly(vinylidene fluoride) (PVDF) culture supports were prepared with structures ranging between 2D and 3D, based on PVDF films on which PVDF microspheres were deposited with varying surface density. Maintenance of multipotentiality when cultured in expansion medium was studied by flow cytometry monitoring the expression of characteristic hMSCs markers, and revealed that cells were losing their characteristic surface markers on these supports. Cell morphology was assessed by scanning electron microscopy (SEM). Alkaline phosphatase activity was also assessed after seven days of culture on expansion medium. On the other hand, osteoblastic differentiation was monitored while culturing in osteogenic medium after cells reached confluence. Osteocalcin immunocytochemistry and alizarin red assays were performed. We show that flow cytometry is a suitable technique for the study of the differentiation of hMSC seeded onto biomaterials, giving a quantitative reliable analysis of hMSC-associated markers. We also show that electrosprayed piezoelectric poly(vinylidene fluoride) is a suitable support for tissue engineering purposes, as hMSCs can proliferate, be viable and undergo osteogenic differentiation when chemically stimulated. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Graphical abstract

17125 KiB  
Article
Preparation and Characterization of Resorbable Bacterial Cellulose Membranes Treated by Electron Beam Irradiation for Guided Bone Regeneration
by Sung-Jun An, So-Hyoun Lee, Jung-Bo Huh, Sung In Jeong, Jong-Seok Park, Hui-Jeong Gwon, Eun-Sook Kang, Chang-Mo Jeong and Youn-Mook Lim
Int. J. Mol. Sci. 2017, 18(11), 2236; https://doi.org/10.3390/ijms18112236 - 25 Oct 2017
Cited by 42 | Viewed by 5685
Abstract
Bacterial cellulose (BC) is an excellent biomaterial with many medical applications. In this study, resorbable BC membranes were prepared for guided bone regeneration (GBR) using an irradiation technique for applications in the dental field. Electron beam irradiation (EI) increases biodegradation by severing the [...] Read more.
Bacterial cellulose (BC) is an excellent biomaterial with many medical applications. In this study, resorbable BC membranes were prepared for guided bone regeneration (GBR) using an irradiation technique for applications in the dental field. Electron beam irradiation (EI) increases biodegradation by severing the glucose bonds of BC. BC membranes irradiated at 100 kGy or 300 kGy were used to determine optimal electron beam doses. Electron beam irradiated BC membranes (EI-BCMs) were evaluated by scanning electron microscopy (SEM), attenuated total reflectance-Fourier transform infrared (ATR-FTIR) spectroscopy, thermal gravimetric analysis (TGA), and using wet tensile strength measurements. In addition, in vitro cell studies were conducted in order to confirm the cytocompatibility of EI-BCMs. Cell viabilities of NIH3T3 cells on 100k and 300k EI-BCMs (100 kGy and 300 kGy irradiated BC membranes) were significantly greater than on NI-BCMs after 3 and 7 days (p < 0.05). Bone regeneration by EI-BCMs and their biodegradabilities were also evaluated using in vivo rat calvarial defect models for 4 and 8 weeks. Histometric results showed 100k EI-BCMs exhibited significantly larger new bone area (NBA; %) than 300k EI-BCMs at 8 weeks after implantation (p < 0.05). Mechanical, chemical, and biological analyses showed EI-BCMs effectively interacted with cells and promoted bone regeneration. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Graphical abstract

4407 KiB  
Article
Collagen-Based Medical Device as a Stem Cell Carrier for Regenerative Medicine
by Léa Aubert, Marie Dubus, Hassan Rammal, Camille Bour, Céline Mongaret, Camille Boulagnon-Rombi, Roselyne Garnotel, Céline Schneider, Rachid Rahouadj, Cedric Laurent, Sophie C. Gangloff, Frédéric Velard, Cedric Mauprivez and Halima Kerdjoudj
Int. J. Mol. Sci. 2017, 18(10), 2210; https://doi.org/10.3390/ijms18102210 - 21 Oct 2017
Cited by 18 | Viewed by 6058
Abstract
Maintenance of mesenchymal stem cells (MSCs) requires a tissue-specific microenvironment (i.e., niche), which is poorly represented by the typical plastic substrate used for two-dimensional growth of MSCs in a tissue culture flask. The objective of this study was to address the potential use [...] Read more.
Maintenance of mesenchymal stem cells (MSCs) requires a tissue-specific microenvironment (i.e., niche), which is poorly represented by the typical plastic substrate used for two-dimensional growth of MSCs in a tissue culture flask. The objective of this study was to address the potential use of collagen-based medical devices (HEMOCOLLAGENE®, Saint-Maur-des-Fossés, France) as mimetic niche for MSCs with the ability to preserve human MSC stemness in vitro. With a chemical composition similar to type I collagen, HEMOCOLLAGENE® foam presented a porous and interconnected structure (>90%) and a relative low elastic modulus of around 60 kPa. Biological studies revealed an apparently inert microenvironment of HEMOCOLLAGENE® foam, where 80% of cultured human MSCs remained viable, adopted a flattened morphology, and maintained their undifferentiated state with basal secretory activity. Thus, three-dimensional HEMOCOLLAGENE® foams present an in vitro model that mimics the MSC niche with the capacity to support viable and quiescent MSCs within a low stiffness collagen I scaffold simulating Wharton’s jelly. These results suggest that haemostatic foam may be a useful and versatile carrier for MSC transplantation for regenerative medicine applications. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Graphical abstract

11152 KiB  
Article
Poly(Lactic Acid) Blends with Poly(Trimethylene Carbonate) as Biodegradable Medical Adhesive Material
by Shuang Zhang, Hongli Li, Mingwei Yuan, Minglong Yuan and Haiyun Chen
Int. J. Mol. Sci. 2017, 18(10), 2041; https://doi.org/10.3390/ijms18102041 - 28 Sep 2017
Cited by 27 | Viewed by 5373
Abstract
A novel medical adhesive was prepared by blending poly(lactic acid) (PLA) with poly(trimethylene carbonate) (PTMC) in ethyl acetate, and the two materials were proven to be biodegradable and biocompatible. The medical adhesive was characterized by 1H nuclear magnetic resonance (1HNMR), [...] Read more.
A novel medical adhesive was prepared by blending poly(lactic acid) (PLA) with poly(trimethylene carbonate) (PTMC) in ethyl acetate, and the two materials were proven to be biodegradable and biocompatible. The medical adhesive was characterized by 1H nuclear magnetic resonance (1HNMR), gel permeation chromatography (GPC), scanning electron microscopy (SEM) and differential scanning calorimetry (DSC). The water vapor transmission rate (WVTR) of this material was measured to be 7.13 g·cm−2·24 h−1. Its degree of comfortability was confirmed by the extensibility (E) and the permanent set (PS), which were approximately 7.83 N·cm−2 and 18.83%, respectively. In vivo tests regarding rabbit immunoglobulin M (IgM), rabbit immunoglobulin G (IgG), rabbit bone alkaline phosphatase (BALP), rabbit interleukin 6 (IL-6), rabbit interleukin 10 (IL-10), rabbit tumor necrosis factor α(TNFα), glutamic-oxaloacetic transaminase (AST/GOT), glutamic-pyruvic transaminase (ALT/GPT), alkaline phosphatase (AKP), blood urea nitrogen (BUN) and creatinine (Cr) indicated that the PLA-PTMC medical adhesive was not harmful to the liver and kidneys. Finally, pathological sections indicated that PLA-PTMC was more effective than the control group. These data suggest that in addition to having a positive effect on hemostasis and no sensibility to wounds, PLA-PTMC can efficiently prevent infections and has great potential as a medical adhesive. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Graphical abstract

3099 KiB  
Article
3D Printed, Microgroove Pattern-Driven Generation of Oriented Ligamentous Architectures
by Chan Ho Park, Kyoung-Hwa Kim, Yong-Moo Lee, William V. Giannobile and Yang-Jo Seol
Int. J. Mol. Sci. 2017, 18(9), 1927; https://doi.org/10.3390/ijms18091927 - 08 Sep 2017
Cited by 35 | Viewed by 5752
Abstract
Specific orientations of regenerated ligaments are crucially required for mechanoresponsive properties and various biomechanical adaptations, which are the key interplay to support mineralized tissues. Although various 2D platforms or 3D printing systems can guide cellular activities or aligned organizations, it remains a challenge [...] Read more.
Specific orientations of regenerated ligaments are crucially required for mechanoresponsive properties and various biomechanical adaptations, which are the key interplay to support mineralized tissues. Although various 2D platforms or 3D printing systems can guide cellular activities or aligned organizations, it remains a challenge to develop ligament-guided, 3D architectures with the angular controllability for parallel, oblique or perpendicular orientations of cells required for biomechanical support of organs. Here, we show the use of scaffold design by additive manufacturing for specific topographies or angulated microgroove patterns to control cell orientations such as parallel (0°), oblique (45°) and perpendicular (90°) angulations. These results demonstrate that ligament cells displayed highly predictable and controllable orientations along microgroove patterns on 3D biopolymeric scaffolds. Our findings demonstrate that 3D printed topographical approaches can regulate spatiotemporal cell organizations that offer strong potential for adaptation to complex tissue defects to regenerate ligament-bone complexes. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Graphical abstract

2663 KiB  
Article
In Situ Forming Gelatin Hydrogels-Directed Angiogenic Differentiation and Activity of Patient-Derived Human Mesenchymal Stem Cells
by Yunki Lee, Daniel A. Balikov, Jung Bok Lee, Sue Hyun Lee, Seung Hwan Lee, Jong Hun Lee, Ki Dong Park and Hak-Joon Sung
Int. J. Mol. Sci. 2017, 18(8), 1705; https://doi.org/10.3390/ijms18081705 - 04 Aug 2017
Cited by 16 | Viewed by 5549
Abstract
Directing angiogenic differentiation of mesenchymal stem cells (MSCs) still remains challenging for successful tissue engineering. Without blood vessel formation, stem cell-based approaches are unable to fully regenerate damaged tissues due to limited support for cell viability and desired tissue/organ functionality. Herein, we report [...] Read more.
Directing angiogenic differentiation of mesenchymal stem cells (MSCs) still remains challenging for successful tissue engineering. Without blood vessel formation, stem cell-based approaches are unable to fully regenerate damaged tissues due to limited support for cell viability and desired tissue/organ functionality. Herein, we report in situ cross-linkable gelatin−hydroxyphenyl propionic acid (GH) hydrogels that can induce pro-angiogenic profiles of MSCs via purely material-driven effects. This hydrogel directed endothelial differentiation of mouse and human patient-derived MSCs through integrin-mediated interactions at the cell-material interface, thereby promoting perfusable blood vessel formation in vitro and in vivo. The causative roles of specific integrin types (α1 and αvβ3) in directing endothelial differentiation were verified by blocking the integrin functions with chemical inhibitors. In addition, to verify the material-driven effect is not species-specific, we confirmed in vitro endothelial differentiation and in vivo blood vessel formation of patient-derived human MSCs by this hydrogel. These findings provide new insight into how purely material-driven effects can direct endothelial differentiation of MSCs, thereby promoting vascularization of scaffolds towards tissue engineering and regenerative medicine applications in humans. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Graphical abstract

Review

Jump to: Editorial, Research, Other

20 pages, 91191 KiB  
Review
Biomimetic Layer-by-Layer Self-Assembly of Nanofilms, Nanocoatings, and 3D Scaffolds for Tissue Engineering
by Shichao Zhang, Malcolm Xing and Bingyun Li
Int. J. Mol. Sci. 2018, 19(6), 1641; https://doi.org/10.3390/ijms19061641 - 01 Jun 2018
Cited by 66 | Viewed by 11942
Abstract
Achieving surface design and control of biomaterial scaffolds with nanometer- or micrometer-scaled functional films is critical to mimic the unique features of native extracellular matrices, which has significant technological implications for tissue engineering including cell-seeded scaffolds, microbioreactors, cell assembly, tissue regeneration, etc. Compared [...] Read more.
Achieving surface design and control of biomaterial scaffolds with nanometer- or micrometer-scaled functional films is critical to mimic the unique features of native extracellular matrices, which has significant technological implications for tissue engineering including cell-seeded scaffolds, microbioreactors, cell assembly, tissue regeneration, etc. Compared with other techniques available for surface design, layer-by-layer (LbL) self-assembly technology has attracted extensive attention because of its integrated features of simplicity, versatility, and nanoscale control. Here we present a brief overview of current state-of-the-art research related to the LbL self-assembly technique and its assembled biomaterials as scaffolds for tissue engineering. An overview of the LbL self-assembly technique, with a focus on issues associated with distinct routes and driving forces of self-assembly, is described briefly. Then, we highlight the controllable fabrication, properties, and applications of LbL self-assembly biomaterials in the forms of multilayer nanofilms, scaffold nanocoatings, and three-dimensional scaffolds to systematically demonstrate advances in LbL self-assembly in the field of tissue engineering. LbL self-assembly not only provides advances for molecular deposition but also opens avenues for the design and development of innovative biomaterials for tissue engineering. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Graphical abstract

15 pages, 2603 KiB  
Review
Recent Advances in Laser-Ablative Synthesis of Bare Au and Si Nanoparticles and Assessment of Their Prospects for Tissue Engineering Applications
by Ahmed Al-Kattan, Viraj P. Nirwan, Anton Popov, Yury V. Ryabchikov, Gleb Tselikov, Marc Sentis, Amir Fahmi and Andrei V. Kabashin
Int. J. Mol. Sci. 2018, 19(6), 1563; https://doi.org/10.3390/ijms19061563 - 24 May 2018
Cited by 32 | Viewed by 5402
Abstract
Driven by surface cleanness and unique physical, optical and chemical properties, bare (ligand-free) laser-synthesized nanoparticles (NPs) are now in the focus of interest as promising materials for the development of advanced biomedical platforms related to biosensing, bioimaging and therapeutic drug delivery. We recently [...] Read more.
Driven by surface cleanness and unique physical, optical and chemical properties, bare (ligand-free) laser-synthesized nanoparticles (NPs) are now in the focus of interest as promising materials for the development of advanced biomedical platforms related to biosensing, bioimaging and therapeutic drug delivery. We recently achieved significant progress in the synthesis of bare gold (Au) and silicon (Si) NPs and their testing in biomedical tasks, including cancer imaging and therapy, biofuel cells, etc. We also showed that these nanomaterials can be excellent candidates for tissue engineering applications. This review is aimed at the description of our recent progress in laser synthesis of bare Si and Au NPs and their testing as functional modules (additives) in innovative scaffold platforms intended for tissue engineering tasks. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Graphical abstract

14 pages, 3295 KiB  
Review
Electrospun Fibrous Scaffolds for Tissue Engineering: Viewpoints on Architecture and Fabrication
by Indong Jun, Hyung-Seop Han, James R. Edwards and Hojeong Jeon
Int. J. Mol. Sci. 2018, 19(3), 745; https://doi.org/10.3390/ijms19030745 - 06 Mar 2018
Cited by 313 | Viewed by 12184
Abstract
Electrospinning has been used for the fabrication of extracellular matrix (ECM)-mimicking fibrous scaffolds for several decades. Electrospun fibrous scaffolds provide nanoscale/microscale fibrous structures with interconnecting pores, resembling natural ECM in tissues, and showing a high potential to facilitate the formation of artificial functional [...] Read more.
Electrospinning has been used for the fabrication of extracellular matrix (ECM)-mimicking fibrous scaffolds for several decades. Electrospun fibrous scaffolds provide nanoscale/microscale fibrous structures with interconnecting pores, resembling natural ECM in tissues, and showing a high potential to facilitate the formation of artificial functional tissues. In this review, we summarize the fundamental principles of electrospinning processes for generating complex fibrous scaffold geometries that are similar in structural complexity to the ECM of living tissues. Moreover, several approaches for the formation of three-dimensional fibrous scaffolds arranged in hierarchical structures for tissue engineering are also presented. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Figure 1

26 pages, 9445 KiB  
Review
Electrospinning of Chitosan-Based Solutions for Tissue Engineering and Regenerative Medicine
by Saad B. Qasim, Muhammad S. Zafar, Shariq Najeeb, Zohaib Khurshid, Altaf H. Shah, Shehriar Husain and Ihtesham Ur Rehman
Int. J. Mol. Sci. 2018, 19(2), 407; https://doi.org/10.3390/ijms19020407 - 30 Jan 2018
Cited by 225 | Viewed by 13725
Abstract
Electrospinning has been used for decades to generate nano-fibres via an electrically charged jet of polymer solution. This process is established on a spinning technique, using electrostatic forces to produce fine fibres from polymer solutions. Amongst, the electrospinning of available biopolymers (silk, cellulose, [...] Read more.
Electrospinning has been used for decades to generate nano-fibres via an electrically charged jet of polymer solution. This process is established on a spinning technique, using electrostatic forces to produce fine fibres from polymer solutions. Amongst, the electrospinning of available biopolymers (silk, cellulose, collagen, gelatine and hyaluronic acid), chitosan (CH) has shown a favourable outcome for tissue regeneration applications. The aim of the current review is to assess the current literature about electrospinning chitosan and its composite formulations for creating fibres in combination with other natural polymers to be employed in tissue engineering. In addition, various polymers blended with chitosan for electrospinning have been discussed in terms of their potential biomedical applications. The review shows that evidence exists in support of the favourable properties and biocompatibility of chitosan electrospun composite biomaterials for a range of applications. However, further research and in vivo studies are required to translate these materials from the laboratory to clinical applications. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Figure 1

24 pages, 2892 KiB  
Review
Tissue Engineering to Improve Immature Testicular Tissue and Cell Transplantation Outcomes: One Step Closer to Fertility Restoration for Prepubertal Boys Exposed to Gonadotoxic Treatments
by Federico Del Vento, Maxime Vermeulen, Francesca De Michele, Maria Grazia Giudice, Jonathan Poels, Anne Des Rieux and Christine Wyns
Int. J. Mol. Sci. 2018, 19(1), 286; https://doi.org/10.3390/ijms19010286 - 18 Jan 2018
Cited by 47 | Viewed by 7174
Abstract
Despite their important contribution to the cure of both oncological and benign diseases, gonadotoxic therapies present the risk of a severe impairment of fertility. Sperm cryopreservation is not an option to preserve prepubertal boys’ reproductive potential, as their seminiferous tubules only contain spermatogonial [...] Read more.
Despite their important contribution to the cure of both oncological and benign diseases, gonadotoxic therapies present the risk of a severe impairment of fertility. Sperm cryopreservation is not an option to preserve prepubertal boys’ reproductive potential, as their seminiferous tubules only contain spermatogonial stem cells (as diploid precursors of spermatozoa). Cryobanking of human immature testicular tissue (ITT) prior to gonadotoxic therapies is an accepted practice. Evaluation of cryopreserved ITT using xenotransplantation in nude mice showed the survival of a limited proportion of spermatogonia and their ability to proliferate and initiate differentiation. However, complete spermatogenesis could not be achieved in the mouse model. Loss of germ cells after ITT grafting points to the need to optimize the transplantation technique. Tissue engineering, a new branch of science that aims at improving cellular environment using scaffolds and molecules administration, might be an approach for further progress. In this review, after summarizing the lessons learned from human prepubertal testicular germ cells or tissue xenotransplantation experiments, we will focus on the benefits that might be gathered using bioengineering techniques to enhance transplantation outcomes by optimizing early tissue graft revascularization, protecting cells from toxic insults linked to ischemic injury and exploring strategies to promote cellular differentiation. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Graphical abstract

4307 KiB  
Review
Ionic Substitutions in Non-Apatitic Calcium Phosphates
by Aleksandra Laskus and Joanna Kolmas
Int. J. Mol. Sci. 2017, 18(12), 2542; https://doi.org/10.3390/ijms18122542 - 27 Nov 2017
Cited by 54 | Viewed by 7958
Abstract
Calcium phosphate materials (CaPs) are similar to inorganic part of human mineralized tissues (i.e., bone, enamel, and dentin). Owing to their high biocompatibility, CaPs, mainly hydroxyapatite (HA), have been investigated for their use in various medical applications. One of the most widely used [...] Read more.
Calcium phosphate materials (CaPs) are similar to inorganic part of human mineralized tissues (i.e., bone, enamel, and dentin). Owing to their high biocompatibility, CaPs, mainly hydroxyapatite (HA), have been investigated for their use in various medical applications. One of the most widely used ways to improve the biological and physicochemical properties of HA is ionic substitution with trace ions. Recent developments in bioceramics have already demonstrated that introducing foreign ions is also possible in other CaPs, such as tricalcium phosphates (amorphous as well as α and β crystalline forms) and brushite. The purpose of this paper is to review recent achievements in the field of non-apatitic CaPs substituted with various ions. Particular attention will be focused on tricalcium phosphates (TCP) and “additives” such as magnesium, zinc, strontium, and silicate ions, all of which have been widely investigated thanks to their important biological role. This review also highlights some of the potential biomedical applications of non-apatitic substituted CaPs. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Graphical abstract

Other

4063 KiB  
Opinion
Designing Smart Biomaterials for Tissue Engineering
by Ferdous Khan and Masaru Tanaka
Int. J. Mol. Sci. 2018, 19(1), 17; https://doi.org/10.3390/ijms19010017 - 21 Dec 2017
Cited by 188 | Viewed by 13156
Abstract
The engineering of human tissues to cure diseases is an interdisciplinary and a very attractive field of research both in academia and the biotechnology industrial sector. Three-dimensional (3D) biomaterial scaffolds can play a critical role in the development of new tissue morphogenesis via [...] Read more.
The engineering of human tissues to cure diseases is an interdisciplinary and a very attractive field of research both in academia and the biotechnology industrial sector. Three-dimensional (3D) biomaterial scaffolds can play a critical role in the development of new tissue morphogenesis via interacting with human cells. Although simple polymeric biomaterials can provide mechanical and physical properties required for tissue development, insufficient biomimetic property and lack of interactions with human progenitor cells remain problematic for the promotion of functional tissue formation. Therefore, the developments of advanced functional biomaterials that respond to stimulus could be the next choice to generate smart 3D biomimetic scaffolds, actively interacting with human stem cells and progenitors along with structural integrity to form functional tissue within a short period. To date, smart biomaterials are designed to interact with biological systems for a wide range of biomedical applications, from the delivery of bioactive molecules and cell adhesion mediators to cellular functioning for the engineering of functional tissues to treat diseases. Full article
(This article belongs to the Special Issue Novel Biomaterials for Tissue Engineering 2018)
Show Figures

Figure 1

Back to TopTop