Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 127
1.
ACS Biomater Sci Eng ; 10(4): 2001-2021, 2024 Apr 08.
Article En | MEDLINE | ID: mdl-38487853

Despite the advances in tissue engineering approaches, reconstruction of long segmental peripheral nerve defects remains unsatisfactory. Although autologous grafts with proper fascicular complementation have shown meaningful functional recovery according to the Medical Research Council Classification (MRCC), the lack of donor nerve for such larger defect sizes (>30 mm) has been a serious clinical issue. Further clinical use of hollow nerve conduits is limited to bridging smaller segmental defects of denuded nerve ends (<30 mm). Recently, bioinspired multichannel nerve guidance conduits (NGCs) gained attention as autograft substitutes as they mimic the fascicular connective tissue microarchitecture in promoting aligned axonal outgrowth with desirable innervation for complete sensory and motor function restoration. This review outlines the hierarchical organization of nerve bundles and their significance in the sensory and motor functions of peripheral nerves. This review also emphasizes the major challenges in addressing the longer nerve defects with the role of fascicular arrangement in the multichannel nerve guidance conduits and the need for fascicular matching to accomplish complete functional restoration, especially in treating long segmental nerve defects. Further, currently available fabrication strategies in developing multichannel nerve conduits and their inconsistency in existing preclinical outcomes captured in this review would seed a new process in designing an ideal larger nerve conduit for peripheral nerve repair.


Peripheral Nerve Injuries , Humans , Peripheral Nerve Injuries/surgery , Sciatic Nerve/injuries , Sciatic Nerve/physiology , Sciatic Nerve/surgery , Tissue Scaffolds , Tissue Engineering
2.
Int J Biol Macromol ; 264(Pt 1): 130565, 2024 Apr.
Article En | MEDLINE | ID: mdl-38432268

Healing chronic and critical-sized full-thickness wounds is a major challenge in the healthcare sector. Scaffolds prepared using electrospinning and hydrogels serve as effective treatment options for wound healing by mimicking the native skin microenvironment. Combining synthetic nanofibers with tunable hydrogel properties can effectively overcome limitations in skin scaffolds made only with nanofibers or hydrogels. In this study, a biocompatible hybrid scaffold was developed for wound healing applications using poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV) nanofibers embedded with hydrogel made of 2 % carboxymethyl cellulose (CMC) blended with 1 % agarose. Hybrid scaffolds, characterized for surface morphology, swellability, porosity, and degradation, were found to be suitable for wound healing. Furthermore, the incorporation of CMC-agarose hydrogel into nanofibers significantly enhanced their mechanical strength compared to PHBV nanofibers alone (p < 0.05). Extract cytotoxicity and direct cytotoxicity tests showed that the hybrid scaffolds developed in this study are cytocompatible (>75 % viability). Furthermore, human adult dermal fibroblasts (HDFa) and human adult immortalized keratinocytes (HaCaT) adhesion, viability, and proliferation studies revealed that the hybrid scaffolds exhibited a significant increase in cell proliferation over time, similar to PHBV nanofibers. Finally, the developed hybrid scaffolds were evaluated in rat full-thickness wounds, demonstrating their ability to promote full-thickness wound healing with reepithelialization and epidermis closure.


Nanofibers , Polyhydroxybutyrates , Tissue Scaffolds , Rats , Humans , Animals , Carboxymethylcellulose Sodium , Sepharose , Skin Transplantation , Hydrogels/pharmacology , Polyesters , Hydroxybutyrates
3.
Int J Biol Macromol ; 260(Pt 1): 129443, 2024 Mar.
Article En | MEDLINE | ID: mdl-38228200

3D bioprinting has emerged as a viable tool to fabricate 3D tissue constructs with high precision using various bioinks which offer instantaneous gelation, shape fidelity, and cytocompatibility. Among various bioinks, cellulose is the most abundantly available natural polymer & widely used as bioink for 3D bioprinting applications. To mitigate the demanding crosslinking needs of cellulose, it is frequently chemically modified or blended with other polymers to develop stable hydrogels. In this study, we have developed a thermoresponsive, composite bioink using carboxymethyl cellulose (CMC) and agarose in different ratios (9:1, 8:2, 7:3, 6:4, and 5:5). Among the tested combinations, the 5:5 ratio showed better gel formation at 37 °C and were further characterized for physicochemical properties. Cytocompatibility was assessed by in vitro extract cytotoxicity assay (ISO 10993-5) using skin fibroblasts cells. CMC-agarose (5:5) bioink was successfully used to fabricate complex 3D structures through extrusion bioprinting and maintained over 80 % cell viability over seven days. Finally, in vivo studies using rat full-thickness wounds showed the potential of CMC-agarose bulk and bioprinted gels in promoting skin regeneration. These results indicate the cytocompatibility and suitability of CMC-agarose bioinks for tissue engineering and 3D bioprinting applications.


Bioprinting , Regenerative Medicine , Rats , Animals , Sepharose , Carboxymethylcellulose Sodium , Printing, Three-Dimensional , Tissue Engineering/methods , Hydrogels/pharmacology , Hydrogels/chemistry , Cellulose/pharmacology , Cellulose/chemistry , Bioprinting/methods , Tissue Scaffolds/chemistry
4.
J Mater Chem B ; 12(2): 350-381, 2024 Jan 03.
Article En | MEDLINE | ID: mdl-38084021

3D printed/bioprinted tissue constructs are utilized for the regeneration of damaged tissues and as in vitro models. Most of the fabricated 3D constructs fail to undergo functional maturation in conventional in vitro settings. There is a challenge to provide a suitable niche for the fabricated tissue constructs to undergo functional maturation. Bioreactors have emerged as a promising tool to enhance tissue maturation of the engineered constructs by providing physical/biological cues along with a controlled nutrient supply under dynamic in vitro conditions. Bioreactors provide an ambient microenvironment most appropriate for the development of functionally matured tissue constructs by promoting cell proliferation, differentiation, and maturation for transplantation and drug screening applications. Due to the huge cost and limited availability of commercial bioreactors, there is a need to develop strategies to make customized bioreactors. Additive manufacturing (AM) may be a viable tool to fabricate custom designed bioreactors with better efficiency and at low cost. In this review, we have extensively discussed the importance of bioreactors in functionalizing tissue engineered/3D bioprinted scaffolds for bone, cartilage, skeletal muscle, nerve, and vascular tissue. In addition, the importance and fabrication of customized 3D printed bioreactors for the maturation of tissue engineered constructs are discussed in detail. Finally, the current challenges and future perspectives in translating commercial and custom 3D printed bioreactors for clinical applications are outlined.


Tissue Engineering , Tissue Scaffolds , Cartilage , Bioreactors , Printing, Three-Dimensional
5.
Biomater Adv ; 152: 213486, 2023 Sep.
Article En | MEDLINE | ID: mdl-37302210

Myocardial infarction (MI) is a lethal cardiac disease that causes most of the mortality across the world. MI is a consequence of plaque in the arterial walls of heart, which eventually result in occlusion and ischemia to the myocardial tissues due to inadequate nutrient and oxygen supply. As an efficient alternative to the existing treatment strategies for MI, 3D bioprinting has evolved as an advanced tissue fabrication technique where the cell-laden bioinks are printed layer-by-layer to create functional cardiac patches. In this study, a dual crosslinking strategy has been utilized towards 3D bioprinting of myocardial constructs by using a combination of alginate and fibrinogen. Herein, pre-crosslinking of the physically blended alginate-fibrinogen bioinks with CaCl2 enhanced the shape fidelity and printability of the printed structures. Physicochemical properties of the bioinks such as rheology, fibrin distribution, swelling ratio and degradation behaviour, were determined post-printing for only ionically crosslinked & dual crosslinked constructs and found to be ideal for bioprinting of cardiac constructs. Human ventricular cardiomyocytes (AC 16) exhibited a significant increase in cell proliferation on day 7 and 14 in AF-DMEM-20 mM CaCl2 bioink when compared to A-DMEM-20 mM CaCl2 (p < 0.05). Furthermore, myocardial patches containing neonatal ventricular rat myocytes (NVRM) showed >80 % viability and also expressed sarcomeric alpha actinin & connexin 43. These results indicate that the dual crosslinking strategy was cytocompatible and also possess the potential to be used for biofabrication of thick myocardial constructs for regenerative medicine applications.


Bioprinting , Hemostatics , Rats , Humans , Animals , Calcium Chloride/pharmacology , Printing, Three-Dimensional , Myocardium , Myocytes, Cardiac , Alginates , Bioprinting/methods
6.
SLAS Technol ; 28(3): 183-198, 2023 06.
Article En | MEDLINE | ID: mdl-37149220

Polysaccharide based hydrogels have been predominantly utilized as ink materials for 3D bioprinting due to biocompatibility and cell responsive features. However, most hydrogels require extensive crosslinking due to poor mechanical properties leading to limited printability. To improve printability without using cytotoxic crosslinkers, thermoresponsive bioinks could be developed. Agarose is a thermoresponsive polysaccharide with upper critical solution temperature (UCST) for sol-gel transition at 35-37 °C. Therefore, we hypothesized that a triad of carboxymethyl cellulose(C)-agarose(A)-gelatin(G) could be a suitable thermoresponsive ink for printing since they undergo instantaneous gelation without any addition of crosslinkers after bioprinting. The blend of agarose-carboxymethyl cellulose was mixed with 1% w/v, 3% w/v and 5% w/v gelatin to optimize the triad ratio for hydrogel formation. It was observed that a blend (C2-A0.5-G1 and C2-A1-G1) containing 2% w/v carboxymethyl cellulose, 0.5% or 1% w/v agarose and 1% w/v gelatin formed better hydrogels with higher stability for up to 21 days in DPBS at 37 °C. Further, C2-A0.5-G1 and C2-A1-G1hydrogels showed higher storage modulus 762 ± 182 Pa & 2452 ± 430 Pa, higher porosity of 96.98 ± 2% & 98.2 ± 0.8% and swellability of 1518 ± 68% & 1587 ± 25% respectively. To evaluate the in vitro potential of these bioink formulations, indirect and direct cytotoxicity were determined using NCTC clone 929 (mouse fibroblast cells) and HADF (primary human adult dermal fibroblast) cells as per the ISO 10993-5 standards. Importantly, the printability of these bioinks was confirmed using extrusion bioprinting by successfully printing different complex 3D patterns.


Carboxymethylcellulose Sodium , Gelatin , Mice , Animals , Humans , Sepharose , Printing, Three-Dimensional , Rheology , Hydrogels
7.
Acta Pharm Sin B ; 13(4): 1711-1725, 2023 Apr.
Article En | MEDLINE | ID: mdl-37139411

Circulating tumor clusters (CTC) disseminating from the primary tumor are responsible for secondary tumor formation where the conventional treatments such as chemotherapy and radiotherapy does not prevent the metastasis at locally advanced stage of breast cancer. In this study, a smart nanotheranostic system has been developed to track and eliminate the CTCs before it can colonize at a new site, which would reduce metastatic progression and increase the five-year survival rate of the breast cancer patients. Targeted multiresponsive (magnetic hyperthermia and pH) nanomicelles incorporated with NIR fluorescent superparamagnetic iron oxide nanoparticles were developed based on self-assembly for dual modal imaging and dual toxicity for spontaneous killing of CTCs in blood stream. A heterogenous tumor clusters model was developed to mimic the CTCs isolated from breast cancer patients. The nanotheranostic system was further evaluated for the targeting property, drug release kinetics, hyperthermia and cytotoxicity against developed CTC model in vitro. In vivo model in BALB/c mice equivalent to stage III and IV human metastatic breast cancer was developed to evaluate the biodistribution and therapeutic efficacy of micellar nanotheranostic system. Reduced CTCs in blood stream and low distant organ metastasis after treatment with the nanotheranostic system demonstrates its potential to capture and kill the CTCs that minimize the secondary tumor formation at distant sites.

8.
ACS Biomater Sci Eng ; 9(6): 3134-3159, 2023 06 12.
Article En | MEDLINE | ID: mdl-37115515

Bioprinting is an additive manufacturing technique that focuses on developing living tissue constructs using bioinks. Bioink is crucial in determining the stability of printed patterns, which remains a major challenge in bioprinting. Thus, the choices of bioink composition, modifications, and cross-linking methods are being continuously researched to augment the clinical translation of bioprinted constructs. Hyaluronic acid (HA) is a naturally occurring polysaccharide with the repeating unit of N-acetyl-glucosamine and d-glucuronic acid disaccharides. It is present in the extracellular matrix (ECM) of tissues (skin, cartilage, nerve, muscle, etc.) with a wide range of molecular weights. Due to the nature of its chemical structure, HA could be easily subjected to chemical modifications and cross-linking that would enable better printability and stability. These interesting properties have made HA an ideal choice of bioinks for developing tissue constructs for regenerative medicine applications. In this Review, the physicochemical properties, reaction chemistry involved in various cross-linking strategies, and biomedical applications of HA have been elaborately discussed. Further, the features of HA bioinks, emerging strategies in HA bioink preparations, and their applications in 3D bioprinting have been highlighted. Finally, the current challenges and future perspectives in the clinical translation of HA-based bioinks are outlined.


Bioprinting , Tissue Engineering , Tissue Engineering/methods , Hydrogels/chemistry , Tissue Scaffolds/chemistry , Hyaluronic Acid/pharmacology , Bioprinting/methods
9.
SLAS Technol ; 28(3): 102-126, 2023 06.
Article En | MEDLINE | ID: mdl-37028493

Tissue-engineered nerve guidance conduits (NGCs) are a viable clinical alternative to autografts and allografts and have been widely used to treat peripheral nerve injuries (PNIs). Although these NGCs are successful to some extent, they cannot aid in native regeneration by improving native-equivalent neural innervation or regrowth. Further, NGCs exhibit longer recovery period and high cost limiting their clinical applications. Additive manufacturing (AM) could be an alternative to the existing drawbacks of the conventional NGCs fabrication methods. The emergence of the AM technique has offered ease for developing personalized three-dimensional (3D) neural constructs with intricate features and higher accuracy on a larger scale, replicating the native feature of nerve tissue. This review introduces the structural organization of peripheral nerves, the classification of PNI, and limitations in clinical and conventional nerve scaffold fabrication strategies. The principles and advantages of AM-based techniques, including the combinatorial approaches utilized for manufacturing 3D nerve conduits, are briefly summarized. This review also outlines the crucial parameters, such as the choice of printable biomaterials, 3D microstructural design/model, conductivity, permeability, degradation, mechanical property, and sterilization required to fabricate large-scale additive-manufactured NGCs successfully. Finally, the challenges and future directions toward fabricating the 3D-printed/bioprinted NGCs for clinical translation are also discussed.


Nerve Regeneration , Peripheral Nerves , Nerve Regeneration/physiology , Peripheral Nerves/metabolism , Tissue Engineering/methods , Biocompatible Materials/chemistry , Biocompatible Materials/metabolism
11.
ACS Appl Mater Interfaces ; 14(48): 54111-54126, 2022 Dec 07.
Article En | MEDLINE | ID: mdl-36401830

The use of hydrogels as scaffolds for three-dimensional (3D) cell growth is an active area of research in tissue engineering. Herein, we report the self-assembly of an ultrashort peptide, a tetrapeptide, Asp-Leu-IIe-IIe, the shortest peptide sequence from a highly fibrillogenic protein TDP-43, into the hydrogel. The hydrogel was mechanically strong and highly stable, with storage modulus values in MPa ranges. The hydrogel supported the proliferation and successful differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) in its matrix as assessed by cell viability, calcium deposition, alkaline phosphatase (ALP) activity, and the expression of osteogenic marker gene studies. To check whether the hydrogel supports 3D growth and regeneration in in vivo conditions, a rabbit critical bone defect model was used. Micro-computed tomography (CT) and X-ray analysis demonstrated the formation of mineralized neobone in the defect areas, with significantly higher bone mineralization and relative bone densities in animals treated with the peptide hydrogel compared to nontreated and matrigel treatment groups. The ultrashort peptide-based hydrogel developed in this work holds great potential for its further development as tissue regeneration and/or engineering scaffolds.


Bone Density , Hydrogels , Animals , Rabbits , Hydrogels/pharmacology , X-Ray Microtomography , Peptides/pharmacology
12.
Biomater Adv ; 142: 213135, 2022 Nov.
Article En | MEDLINE | ID: mdl-36215745

Skeletal muscles are essential for body movement, and the loss of motor function due to volumetric muscle loss (VML) limits the mobility of patients. Current therapeutic approaches are insufficient to offer complete functional recovery of muscle damages. Tissue engineering provides viable ways to fabricate scaffolds to regenerate damaged tissues. Hence, tissue engineering options are explored to address existing challenges in the treatment options for muscle regeneration. Electrospinning is a widely employed fabrication technique to make muscle mimetic nanofibrous scaffolds for tissue regeneration. 3D bioprinting has also been utilized to fabricate muscle-like tissues in recent times. This review discusses the anatomy of skeletal muscle, defects, the healing process, and various treatment options for VML. Further, the advanced strategies in electrospinning of natural and synthetic polymers are discussed, along with the recent developments in the fabrication of hybrid scaffolds. Current approaches in 3D bioprinting of skeletal muscle tissues are outlined with special emphasis on the combination of electrospinning and 3D bioprinting towards the development of fully functional muscle constructs. Finally, the current challenges and future perspectives of these convergence techniques are discussed.


Bioprinting , Nanofibers , Humans , Bioprinting/methods , Tissue Scaffolds , Tissue Engineering/methods , Muscle, Skeletal/physiology
13.
Biomed Mater ; 18(1)2022 11 10.
Article En | MEDLINE | ID: mdl-36270604

The incidence of highly aggressive pancreatic cancer is increasing across the globe and is projected to increase to 18.6% by 2050. The mortality rate for this form of cancer is very high and the 5 y relative survival rate is only about 9%-10%. The 3D pancreatic cancer microenvironment exerts a major influence on the poor survival rate. A key factor is the prevention of the penetration of the chemotherapeutic drugs in the three-dimensional (3D) microenvironment leading to the development of chemoresistance which is a major contributor to the survival rates. Hence,in vitrostudies using 3D cultures represent a better approach to understand the effect of therapeutic formulations on the cancer cells when compared to conventional 2D cultures. In the present study, we have explored three different conditions for the development of a 3D pancreatic tumour spheroid model from MiaPaCa-2 and PanC1 cells cultured for 10 days using Matrigel matrix. This optimized spheroid model was employed to evaluate a multi-functional nanotheranostic system fabricated using chitosan nanoparticles co-encapsulated with the chemotherapeutic agent gemcitabine and gold-capped iron oxide nanoparticles for multimodal imaging. The effect of the single and multiple-dose regimens of the theranostic system on the viability of 3D spheroids formed from the two pancreatic cancer cell lines was studied. It was observed that the 3D tumour spheroids cultured for 10 days exhibited resistance towards free gemcitabine drug, unlike the 2D culture. The administration of the multifunctional nanotheranostic system on alternate days effectively reduced the cancer cell viability after five doses to about 20% when compared with other groups. The repeated doses of the nanotheranostic system were found to be more effective than the single dose. Cell line-based differences in internalization of the carrier was also reflected in their response to the nanocarrier with PanC1 showing better sensitivity to the treatment.In vivostudies revealed that the combination of gemcitabine and magnetic field induced hypothermia produced superior regression in cancer when compared with the chemotherapeutic agent alone by a combination of activating the pro-apoptotic pathway and heat-induced necrosis. Our results reveal that this multi-functional system holds promise to overcome the current challenges to treat pancreatic cancers.


Antineoplastic Agents , Pancreatic Neoplasms , Humans , Theranostic Nanomedicine , Spheroids, Cellular/pathology , Cell Line, Tumor , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Tumor Microenvironment , Pancreatic Neoplasms
14.
Biomed Mater ; 17(6)2022 Sep 27.
Article En | MEDLINE | ID: mdl-36099909

Small-diameter arterial conduits with native physiological and biological equivalence continues to be a constant global demand posing critical challenges in fabrication. Advent of various strategies towards mimicking the structural hierarchy of a native blood vessel, often involve complex instrumentation and template-assistance with post-processing complications eventually compromising structural fidelity. In the present research, we report a template-free, facile strategy- '3D wet writing' by peripheral-core differential ionic gelation to fabricate perfusable customizable constructs of any dimension, thickness and length in <5 mins. Dual-crosslinking using di-diol complexation of borax with Alginate- poly (vinyl alcohol) was performed to enhance the stability of fabricated bi-layered tubular constructs (BLT). These fabricated BLTs demonstrated non-linear mechanical characteristics of native blood vessels in withstanding physiological (120/80 mmHg) hemodynamic loading conditions with cyclic strain (5.82 ± 0.88%). The BLTs also ensured adequate longitudinal (0.176 ± 0.03 MPa) & circumferential (0.29 ± 0.012 MPa) tensile strength and burst pressure strength of 353.875 ± 22.69 mmHg. Hemocompatible characteristics of BLT were clearly evident with lower hemolytic index (0.21 ± 0.03%) and maintenance of erythrocyte structural integrity under dynamic conditions. Further, non-thrombogenic and non-inflammatory characteristics of BLTs were confirmed by in-activated platelets and monocytes under dynamic conditions. The developed wet-writing technique exhibited facile integration of layer-specific cells concurrently with the BLT fabrication. The spatial cell-specific expressions of smooth muscle (α-SMA) and endothelial (CD-31) cells in BLT were comparable to native hierarchical cellular organization with the multi-layered medial and mono-layered intimal layers. Further,ex-vivodynamic studies on anastomotic interface between BLT and rat abdominal aorta clearly evidenced the functional efficacy of fabricated BLTs as physiologically relevant small-diameter vascular construct.


Tissue Engineering , Tissue Scaffolds , Alginates , Animals , Biomimetics , Hydrogels , Rats , Tissue Scaffolds/chemistry
15.
Biomater Adv ; 134: 112576, 2022 Mar.
Article En | MEDLINE | ID: mdl-35525748

3D bioprinting has enabled the creation of biomimetic tissue constructs for regenerative medicine and in vitro model systems. Large-scale production of 3D structures at the micron-scale resolution is achieved through bioprinting using custom bioinks. Stability and 3D construct compliance play an important role in offering cells with biomechanical cues that regulate their behavior and enable in vivo implantation. Various crosslinking strategies are developed to stabilize the 3D printed structures and new methodologies are constantly being evaluated to overcome the limitations of the existing methods. Photo-crosslinking has emerged as a simple and elegant technique that offers precise control over the spatiotemporal gelation of bioinks during bioprinting. This article summarizes the use of photo-crosslinking agents and methodology towards optimizing 3D constructs for specific biomedical applications. The article also takes into account various bioinks and photo-crosslinkers in creating stable 3D printed structures that offer bioactivity with desirable physicochemical properties. The current challenges of 3D bioprinting and new directions that can advance the field in its wide applicability to create 3D tissue models to study diseases and organ transplantation are also summarized.


Bioprinting , Biomimetics , Bioprinting/methods , Printing, Three-Dimensional , Regenerative Medicine/methods , Tissue Engineering/methods
16.
J Mater Sci Mater Med ; 33(4): 36, 2022 Apr 09.
Article En | MEDLINE | ID: mdl-35397053

The healing physiology of bone repair and remodeling that occurs after normal fracture is well orchestrated. However, it fails in complex clinical conditions and hence requires augmentation by grafts. In this study, composite nanohydroxyapatite (nHA), poly(hydroxybutyrate) (PHB) and poly(ɛ-caprolactone) (PCL) constituted microspheres sintered three-dimensional scaffold were evaluated in rabbit ulnar segmental defect. A composite scaffold using PHB-PCL-nHA microspheres was developed with protein interface by solvent/non-solvent sintering to provide multiple cues such as biocomposition, cancellous bone equivalent meso-micro multi-scale porosity, and compressive strength. In vitro DNA quantification and alkaline phosphatase (ALP) assays revealed that the protein interfaced composite scaffolds supported osteoblast proliferation and mineralization significantly higher than scaffolds without protein and TCPS (p < 0.05). Scanning electron micrographs of osteoblasts cultured scaffolds demonstrated cell-matrix interaction, cell spreading, colonization and filopodial extension across the porous voids. Cylindrical scaffolds (5 × 10 mm) were implanted following segmental defect (10 mm) in rabbit ulnar bone and compared with untreated control. Radiography (4, 8 and 12 weeks) and µ-computed tomography (12 weeks) analysis showed directional bone tissue formation by bridging defective site in both scaffolds with and without protein interface. Whereas, undesired sclerotic-like tissue formation was observed in control groups from 8 weeks. Histology by hot Stevenel's blue and van Gieson's picrofuchsin staining has confirmed enhanced bone maturation in scaffold groups while presence of osteoids was observed in control after 12 weeks. Thus, the developed composite matrices exhibits osteoinductive, osteoconductive properties and demonstrates its bone regenerative potential owing to its compositional, micro & macro structural and mechanical properties. Graphical abstract.


Tissue Engineering , Tissue Scaffolds , Animals , Bone Regeneration , Osteogenesis , Porosity , Rabbits , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Ulna
17.
Mater Horiz ; 9(4): 1141-1166, 2022 04 04.
Article En | MEDLINE | ID: mdl-35006214

DNA has excellent features such as the presence of functional and targeted molecular recognition motifs, tailorability, multifunctionality, high-precision molecular self-assembly, hydrophilicity, and outstanding biocompatibility. Due to these remarkable features, DNA has emerged as a leading next-generation biomaterial of choice to make hydrogels by self-assembly. In recent times, novel routes for the chemical synthesis of DNA, advances in tailorable designs, and affordable production ways have made DNA as a building block material for various applications. These advanced features have made researchers continuously explore the interesting properties of pure and hybrid DNA for 3D bioprinting and other biomedical applications. This review article highlights the topical advancements in the use of DNA as an ideal bioink for the bioprinting of cell-laden three-dimensional tissue constructs for regenerative medicine applications. Various bioprinting techniques and emerging design approaches such as self-assembly, nucleotide sequence, enzymes, and production cost to use DNA as a bioink for bioprinting applications are described. In addition, various types and properties of DNA hydrogels such as stimuli responsiveness and mechanical properties are discussed. Further, recent progress in the applications of DNA in 3D bioprinting are emphasized. Finally, the current challenges and future perspectives of DNA hydrogels in 3D bioprinting and other biomedical applications are discussed.


Bioprinting , Biocompatible Materials/therapeutic use , Bioprinting/methods , DNA , Printing, Three-Dimensional , Tissue Engineering/methods , Tissue Scaffolds
18.
Cells Tissues Organs ; 211(3): 335-347, 2022.
Article En | MEDLINE | ID: mdl-34058730

Modeling of the human vascular microphysiological system (MPS) has gained attention due to precise prediction of drug response and toxicity during drug screening process. Developing a physiologically equivalent vascular MPS still remains complex as it demands the recapitulation of dynamic structural and biological microenvironment similar to native vasculature. Hence, an ideal MPS would involve developing perfusable 3D in vitro models with multilayered human vascular cells encapsulated in a matrix to regulate the vascular tone resembling the native. Several attempts to model such anatomically accurate physiological and pathological blood vessels often fail to harmonize the essential vascular microenvironment. For instance, conventional microfluidic-based approaches employed for vascular MPS, though offering creation of perfusable channel, do not replicate the vascular hierarchical cellular arrangement due to planar geometry and confluent monolayered cell seeding. Also, recent advances with 3D biofabrication strategies are still limited by fabrication of small-diameter constructs and scalability besides post-processing techniques that indirectly distort the structural integrity of the hydrogel tubular constructs. These existing limitations toward fabricating a relevant vascular MPS demand a facile and mechanically stable construct. Hence, the present study is aimed toward developing a stable viable self-standing perfusable hydrogel construct by a rapid and scalable strategy toward vascular MPS application. The fabricated tubular constructs were found to be structurally stable with end-to-end perfusability exhibiting their potential as self-standing perfusable structures. Also, the construct exhibited nonhemolytic behavior with perfusion of red blood cells inside the luminal channel. The present study evidences creation of a dual-crosslinked stable, viable self-standing hydrogel construct with multilayered homogenous distribution of viable smooth muscle cells throughout the construct, thereby demonstrating its applicability as a promising 3D in vitro vascular microphysiological system.


Hydrogels , Tissue Engineering , Humans , Perfusion , Tissue Engineering/methods , Tissue Scaffolds/chemistry
19.
ACS Omega ; 6(48): 32528-32536, 2021 Dec 07.
Article En | MEDLINE | ID: mdl-34901602

The traditional three-electrode electrochemical system used in the development of biosensors for detecting various biomarkers of interest necessitates the use of bulk electrodes, which precludes the deployment of handheld electrochemical devices in clinical applications. Affordable screen-printed carbon electrodes (SPCEs) modified with functional interfaces are being developed to enhance the sensitivity of a compact sensing system as a whole. In this work, SPCEs were fabricated on an overhead projection (OHP) sheet in three different active areas of 2 × 2, 3 × 3, and 4 × 4 mm2 using a screen printing technique, and then ∼2 nm sized graphene quantum dots (GQDs) were electrodeposited over the SPCE surface to add functionality for the detection of ultralow levels of one of the cardiac biomarkers, C-reactive protein (CRP). The proposed mediator-dependent voltammetric biosensor exhibited good sensitivity, a low detection limit, and a linear range of 2.45 µA ng-1 mL-1 cm-2, 0.036 ng mL-1, and 0.5-10 ng mL-1, respectively. The fabricated SPCE/GQDs/anti-CRP biosensor could rapidly detect CRP in less than 25 s. The intra- and interassays were performed with five sensor strips, which showed a minimum standard deviation of 1.85 and 2.8%, respectively. The SPCE/GQDs/anti-CRP electrode was used to detect CRP concentrations in a ringer lactate solution. Thus, the developed biosensor has all of the characteristics such as rapidity, inexpensive disposable electrodes, miniaturization, and a lower detection limit needed to evolve as a point-of-care (PoC) application.

20.
J Tissue Eng Regen Med ; 15(11): 998-1011, 2021 11.
Article En | MEDLINE | ID: mdl-34551457

Reconstruction of peripheral nervous tissue remains challenging in critical-sized defects due to the lack of Büngner bands from the proximal to the distal nerve ends. Conventional nerve guides fail to bridge the large-sized defect owing to the formation of a thin fibrin cable. Hence, in the present study, an attempt was made to reverse engineer the intricate epi-, peri- and endo-neurial tissues using Fused Deposition Modeling based 3D printing. Bovine serum albumin protein nanoflowers (NF) exhibiting Viburnum opulus 'Roseum' morphology were ingrained into 3D printed constructs without affecting its secondary structure to enhance the axonal guidance from proximal to distal ends of denuded nerve ends. Scanning electron micrographs confirmed the uniform distribution of protein NF in 3D printed constructs. The PC-12 cells cultured on protein ingrained 3D printed scaffolds demonstrated cytocompatibility, improved cell adhesion and extended neuronal projections with significantly higher intensities of NF-200 and tubulin expressions. Further suture-free fixation designed in the current 3D printed construct aids facile implantation of printed conduits to the transected nerve ends. Hence the protein ingrained 3D printed construct would be a promising substitute to treat longer peripheral nerve defects as its structural equivalence of endo- and perineurial organization along with the ingrained protein NF promote the neuronal extension towards the distal ends by minimizing axonal dispersion.


Nerve Tissue/physiology , Tissue Engineering , Animals , Cattle , Cell Adhesion , Cell Differentiation , Cell Survival , Goats , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Nerve Tissue/diagnostic imaging , Neurofilament Proteins/metabolism , PC12 Cells , Printing, Three-Dimensional , Rats , Serum Albumin, Bovine/chemistry , Surface Properties , Sutures , Temperature , Tissue Scaffolds/chemistry , X-Ray Microtomography
...