Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 89
Filtrer
1.
Gels ; 9(12)2023 Nov 23.
Article de Anglais | MEDLINE | ID: mdl-38131909

RÉSUMÉ

Hydrogels are three-dimensional (3D) water-swellable polymeric matrices that are used extensively in tissue engineering and drug delivery. Hydrogels can be conformed into any desirable shape using 3D bio-printing, making them suitable for personalized treatment. Among the different 3D bio-printing techniques, digital light processing (DLP)-based printing offers the advantage of quickly fabricating high resolution structures, reducing the chances of cell damage during the printing process. Here, we have used DLP to 3D bio-print biocompatible gelatin methacrylate (GelMA) scaffolds intended for bone repair. GelMA is biocompatible, biodegradable, has integrin binding motifs that promote cell adhesion, and can be crosslinked easily to form hydrogels. However, GelMA on its own is incapable of promoting bone repair and must be supplemented with pharmaceutical molecules or growth factors, which can be toxic or expensive. To overcome this limitation, we introduced zinc-based metal-organic framework (MOF) nanoparticles into GelMA that can promote osteogenic differentiation, providing safer and more affordable alternatives to traditional methods. Incorporation of this nanoparticle into GelMA hydrogel has demonstrated significant improvement across multiple aspects, including bio-printability, and favorable mechanical properties (showing a significant increase in the compressive modulus from 52.14 ± 19.42 kPa to 128.13 ± 19.46 kPa with the addition of ZIF-8 nanoparticles). The designed nanocomposite hydrogels can also sustain drug (vancomycin) release (maximum 87.52 ± 1.6% cumulative amount) and exhibit a remarkable ability to differentiate human adipose-derived mesenchymal stem cells toward the osteogenic lineage. Furthermore, the formulated MOF-integrated nanocomposite hydrogel offers the unique capability to coat metallic implants intended for bone healing. Overall, the remarkable printability and coating ability displayed by the nanocomposite hydrogel presents itself as a promising candidate for drug delivery, cell delivery and bone tissue engineering applications.

2.
ACS Biomater Sci Eng ; 9(7): 3972-3986, 2023 07 10.
Article de Anglais | MEDLINE | ID: mdl-37378614

RÉSUMÉ

Biobased nanoparticles are at the leading edge of the rapidly developing field of nanomedicine and biotherapeutics. Their unique size, shape, and biophysical properties make them attractive tools for biomedical research, including vaccination, targeted drug delivery, and immune therapy. These nanoparticles are engineered to present native cell receptors and proteins on their surfaces, providing a biomimicking camouflage for therapeutic cargo to evade rapid degradation, immune rejection, inflammation, and clearance. Despite showing promising clinical relevance, commercial implementation of these biobased nanoparticles is yet to be fully realized. In this perspective, we discuss advanced biobased nanoparticle designs used in medical applications, such as cell membrane nanoparticles, exosomes, and synthetic lipid-derived nanoparticles, and highlight their benefits and potential challenges. Moreover, we critically assess the future of preparing such particles using artificial intelligence and machine learning. These advanced computational tools will be able to predict the functional composition and behavior of the proteins and cell receptors present on the nanoparticle surfaces. With more advancement in designing new biobased nanoparticles, this field of research could play a key role in dictating the future rational design of drug transporters, thereby ultimately improving overall therapeutic outcomes.


Sujet(s)
Nanomédecine , Nanoparticules , Intelligence artificielle , Nanoparticules/usage thérapeutique , Systèmes de délivrance de médicaments , Protéines
3.
Mol Pharm ; 20(1): 767-774, 2023 01 02.
Article de Anglais | MEDLINE | ID: mdl-36322617

RÉSUMÉ

Natural polymer-based hydrogels are excellent for encapsulating hydrophilic drugs, but they are mechanically weak and degrade easily. In this communication, we exploit the electrostatic interaction between nanosilicates (nSi) and gelatin methacrylate (GelMA) to form a mechanically tough nanocomposite hydrogel for pharmaceutical drug delivery. These hydrogels, prepared at subzero temperatures to form cryogels, displayed macroporous structures, which favors cell infiltration. The designed tough cryogel also showed a slower rate of degradation. Furthermore, we encapsulated the small molecule metformin and sustained the drug release under physiological conditions. Cryogel-loaded metformin reduced the effect of endothelial cell injury caused by nutrient deprivation in vitro. Finally, we hypothesize that this versatile nanocomposite material will find use in diverse biomedical applications.


Sujet(s)
Hydrogels , Nanoparticules , Hydrogels/composition chimique , Cryogels , Préparations pharmaceutiques , Systèmes de délivrance de médicaments , Gélatine/composition chimique , Nanoparticules/composition chimique
4.
Mater Horiz ; 9(7): 1850-1865, 2022 07 04.
Article de Anglais | MEDLINE | ID: mdl-35485266

RÉSUMÉ

Exploring new avenues for clinical management of chronic wounds holds the key to eliminating socioeconomic burdens and health-related concerns associated with this silent killer. Engineered biomaterials offer great promise for repair and regeneration of chronic wounds because of their ability to deliver therapeutics, protect the wound environment, and support the skin matrices to facilitate tissue growth. This mini review presents recent advances in biomaterial functionalities for enhancing wound healing and demonstrates a move from sub-optimal methods to multi-functionalized treatment approaches. In this context, we discuss the recently reported biomaterial characteristics such as bioadhesiveness, antimicrobial properties, proangiogenic attributes, and anti-inflammatory properties that promote chronic wound healing. In addition, we highlight the necessary mechanical and mass transport properties of such biomaterials. Then, we discuss the characteristic properties of various biomaterial templates, including hydrogels, cryogels, nanomaterials, and biomolecule-functionalized materials. These biomaterials can be microfabricated into various structures, including smart patches, microneedles, electrospun scaffolds, and 3D-bioprinted structures, to advance the field of biomaterial scaffolds for effective wound healing. Finally, we provide an outlook on the future while emphasizing the need for their detailed functional behaviour and inflammatory response studies in a complex in vivo environment for superior clinical outcomes and reduced regulatory hurdles.


Sujet(s)
Matériaux biocompatibles , Nanostructures , Matériaux biocompatibles/usage thérapeutique , Peau , Structures d'échafaudage tissulaires/composition chimique , Cicatrisation de plaie/physiologie
5.
Mater Horiz ; 9(4): 1141-1166, 2022 04 04.
Article de Anglais | MEDLINE | ID: mdl-35006214

RÉSUMÉ

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.


Sujet(s)
Bio-impression , Matériaux biocompatibles/usage thérapeutique , Bio-impression/méthodes , ADN , Impression tridimensionnelle , Ingénierie tissulaire/méthodes , Structures d'échafaudage tissulaires
6.
Adv Healthc Mater ; 11(8): e2102088, 2022 04.
Article de Anglais | MEDLINE | ID: mdl-35032156

RÉSUMÉ

Considerable progress has been made in synthesizing "intelligent", biodegradable hydrogels that undergo rapid changes in physicochemical properties once exposed to external stimuli. These advantageous properties of stimulus-triggered materials make them highly appealing to diverse biomedical applications. Of late, research on the incorporation of light-triggered nanoparticles (NPs) into polymeric hydrogel networks has gained momentum due to their ability to remotely tune hydrogel properties using facile, contact-free approaches, such as adjustment of wavelength and intensity of light source. These multi-functional NPs, in combination with tissue-mimicking hydrogels, are increasingly being used for on-demand drug release, preparing diagnostic kits, and fabricating smart scaffolds. Here, the authors discuss the atomic behavior of different NPs in the presence of light, and critically review the mechanisms by which NPs convert light stimuli into heat energy. Then, they explain how these NPs impact the mechanical properties and rheological behavior of NPs-impregnated hydrogels. Understanding the rheological behavior of nanocomposite hydrogels using different sophisticated strategies, including computer-assisted machine learning, is critical for designing the next generation of drug delivery systems. Next, they highlight the salient strategies that have been used to apply light-induced nanocomposites for diverse biomedical applications and provide an outlook for the further improvement of these NPs-driven light-responsive hydrogels.


Sujet(s)
Hydrogels , Nanoparticules , Systèmes de délivrance de médicaments , Hydrogels/composition chimique , Sciences des matériaux , Nanoparticules/composition chimique , Polymères/composition chimique
7.
Drug Discov Today ; 27(4): 1156-1166, 2022 04.
Article de Anglais | MEDLINE | ID: mdl-34839040

RÉSUMÉ

Diabetes mellitus is a chronic disease characterized by increased blood glucose levels, leading to damage of the nerves blood vessels, subsequently manifesting as organ failures, wounds, or ulcerations. Wounds in patients with diabetes are further complicated because of reduced cytokine responses, infection, poor vascularization, and delayed healing processes. Surface-functionalized and bioengineered nanoparticles (NPs) have recently gained attention as emerging treatment modalities for wound healing in diabetes. Here, we review emerging therapeutic NPs to treat diabetic wounds and highlight their discrete delivery mechanisms and sites of action. We further critically assess the current challenges of these nanoengineered materials for successful clinical translation and discuss their potential for growth in the clinical marketplace.


Sujet(s)
Diabète , Nanoparticules , Diabète/traitement médicamenteux , Humains , Cicatrisation de plaie
8.
Biomater Sci ; 9(19): 6337-6354, 2021 Sep 28.
Article de Anglais | MEDLINE | ID: mdl-34397056

RÉSUMÉ

Three-dimensional (3D) bioprinting is an emerging tissue engineering approach that aims to develop cell or biomolecule-laden, complex polymeric scaffolds with high precision, using hydrogel-based "bioinks". Hydrogels are water-swollen, highly crosslinked polymer networks that are soft, quasi-solid, and can support and protect biological materials. However, traditional hydrogels have weak mechanical properties and cannot retain complex structures. They must be reinforced with physical and chemical manipulations to produce a mechanically resilient bioink. Over the past few years, we have witnessed an increased use of nanoparticles and biological moiety-functionalized nanoparticles to fabricate new bioinks. Nanoparticles of varied size, shape, and surface chemistries can provide a unique solution to this problem primarily because of three reasons: (a) nanoparticles can mechanically reinforce hydrogels through physical and chemical interactions. This can favorably influence the bioink's 3D printability and structural integrity by modulating its rheological, biomechanical, and biochemical properties, allowing greater flexibility to print a wide range of structures; (b) nanoparticles can introduce new bio-functionalities to the hydrogels, which is a key metric of a bioink's performance, influencing both cell-material and cell-cell interactions within the hydrogel; (c) nanoparticles can impart "smart" features to the bioink, making the tissue constructs responsive to external stimuli. Responsiveness of the hydrogel to magnetic field, electric field, pH changes, and near-infrared light can be made possible by the incorporation of nanoparticles. Additionally, bioink polymeric networks with nanoparticles can undergo advanced chemical crosslinking, allowing greater flexibility to print structures with varied biomechanical properties. Taken together, the unique properties of various nanoparticles can help bioprint intricate constructs, bringing the process one step closer to complex tissue structure and organ printing. In this review, we explore the design principles and multifunctional properties of various nanomaterials and nanocomposite hydrogels for potential, primarily extrusion-based bioprinting applications. We illustrate the significance of biocompatibility of the designed nanocomposite hydrogel-based bioink for clinical translation and discuss the different parameters that affect cell fate after cell-nanomaterial interaction. Finally, we critically assess the current challenges of nanoengineering bioinks and provide insight into the future directions of potential hydrogel bioinks in the rapidly evolving field of bioprinting.


Sujet(s)
Bio-impression , Nanoparticules , Hydrogels , Impression tridimensionnelle , Ingénierie tissulaire , Structures d'échafaudage tissulaires
9.
Trends Pharmacol Sci ; 42(10): 813-828, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-34454774

RÉSUMÉ

Vaccines have been used to train the immune system to recognize pathogens, and prevent and treat diseases, such as cancer, for decades. However, there are continuing challenges in their manufacturing, large-scale production, and storage. Some of them also show suboptimal immunogenicity, requiring additional adjuvants and booster doses. As an alternate vaccination strategy, a new class of biomimetic materials with unique functionalities has emerged in recent years. Here, we explore the current bioengineering techniques that make use of hydrogels, modified polymers, cell membranes, self-assembled proteins, virus-like particles (VLPs), and nucleic acids to deliver and develop biomaterial-based vaccines. We also review design principles and key regulatory issues associated with their development. Finally, we critically assess their limitations, explore approaches to overcome these limitations, and discuss potential future applications for clinical translation.


Sujet(s)
Matériaux biomimétiques , Vaccins , Matériaux biocompatibles , Hydrogels , Polymères
10.
J Biomed Mater Res A ; 109(12): 2597-2610, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34189837

RÉSUMÉ

Hydrogels can be fabricated and designed to exert direct control over stem cells' adhesion and differentiation. In this study, we have investigated the use of polydopamine (pDA)-treatment as a binding platform for bioactive compounds to create a versatile gelatin-alginate (Gel-Alg) hydrogel for tissue engineering applications. Precisely, pDA was used to modify the surface properties of the hydrogel and better control the adhesion and osteogenic differentiation of human adipose-derived stem cells (hASCs). pDA enabled the adsorption of different types of bioactive molecules, including a model osteoinductive drug (dexamethasone) as well as a model pro-angiogenic peptide (QK). The pDA treatment efficiently retained the drug and the peptide compared to the untreated hydrogel and proved to be effective in controlling the morphology, cell area, and osteogenic differentiation of hASCs. Overall, the findings of this study confirm the efficacy of pDA treatment as a valuable strategy to modulate the biological properties of biocompatible Gel-Alg hydrogels and further extend their value in regenerative medicine.


Sujet(s)
Tissu adipeux/physiologie , Alginates/composition chimique , Gélatine/composition chimique , Hydrogels/composition chimique , Indoles/composition chimique , Polymères/composition chimique , Cellules souches/physiologie , Adhérence cellulaire , Différenciation cellulaire/effets des médicaments et des substances chimiques , Matériaux revêtus, biocompatibles , Dexaméthasone/pharmacologie , Humains , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Ostéogenèse , Médecine régénérative/méthodes , Ingénierie tissulaire , Structures d'échafaudage tissulaires
11.
Chem Soc Rev ; 50(13): 7779-7819, 2021 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-34036968

RÉSUMÉ

The biological purpose of DNA is to store, replicate, and convey genetic information in cells. Progress in molecular genetics have led to its widespread applications in gene editing, gene therapy, and forensic science. However, in addition to its role as a genetic material, DNA has also emerged as a nongenetic, generic material for diverse biomedical applications. DNA is essentially a natural biopolymer that can be precisely programed by simple chemical modifications to construct materials with desired mechanical, biological, and structural properties. This review critically deciphers the chemical tools and strategies that are currently being employed to harness the nongenetic functions of DNA. Here, the primary product of interest has been crosslinked, hydrated polymers, or hydrogels. State-of-the-art applications of macroscopic, DNA-based hydrogels in the fields of environment, electrochemistry, biologics delivery, and regenerative therapy have been extensively reviewed. Additionally, the review encompasses the status of DNA as a clinically and commercially viable material and provides insight into future possibilities.


Sujet(s)
Matériaux biocompatibles/composition chimique , ADN/composition chimique , Hydrogels/composition chimique , Polymères/composition chimique
12.
Nanoscale ; 12(47): 24273-24284, 2020 Dec 21.
Article de Anglais | MEDLINE | ID: mdl-33295935

RÉSUMÉ

Prevailing drug delivery strategies rely on the use of synthetic nanocarriers like metal nanoparticles and polymeric liposomes to control the release of therapeutics in a safe and efficacious manner. Despite their high efficiency in encapsulating drugs, these systems exhibit low to moderate biocompatibility, low cellular uptake, and sub-optimal targeting capabilities. Conversely, cell-derived nanoparticles (CDNs) have emerged as a promising alternative to these artificial drug delivery carriers for achieving safer clinical outcomes. In this study, we have generated CDNs from human adipose-derived stem cells (hASCs) using a high-yield fabrication strategy. Briefly, hASCs were subjected to a cell-shearing approach that entails passing the cells through an array of filters, along with serial centrifugations to eliminate intracellular contents. Ultimately, the fragmented parent cell membrane self-assembles to form the CDNs. This strategy successfully converted 80% of the plasma membrane into the novel nanocarriers with an average hydrodynamic diameter of 100 nm. Stability analysis confirmed that the formulated nanocarriers are stable for over 3 weeks, making them a potent candidate for long-term therapies. To demonstrate their potential in drug delivery, we encapsulated trehalose, a cell-impermeable sugar molecule, into the CDNs via an extrusion loading technique. Drug-loaded CDNs were effectively internalized into human umbilical vein endothelial cells (HUVECs) and hASCs, without inducing any significant cytotoxicity. Overall, the findings of this study establish the potential of hASC-derived CDNs as customizable biomimetic nanocarriers for drug delivery and other translational medicine applications.


Sujet(s)
Systèmes de délivrance de médicaments , Nanoparticules , Biomimétique , Vecteurs de médicaments , Humains , Cellules souches
13.
Acta Biomater ; 105: 159-169, 2020 03 15.
Article de Anglais | MEDLINE | ID: mdl-31972367

RÉSUMÉ

Injectable hydrogels represent a valuable tool for the delivery of therapeutic molecules aimed to restore the functionality of damaged tissues. In this study, we report the design of a nanocomposite DNA-based hydrogel crosslinked with oxidized alginate (OA) via the formation of reversible imine linkages. The formulated hydrogel functioned as an injectable carrier for the sustained delivery of a small molecule drug, simvastatin. The degree of oxidation of alginate and the concentration of silicate-based nanoparticles (nSi) were varied to modulate the rheological properties of the hydrogels. Specifically, the formulations consisting of OA with higher degree of oxidation displayed the highest value of storage moduli, yield stress, yield strain, and rapid recovery after removal of cyclic stress. The hydrogel formulations exhibited self-healing and shear-thinning properties due to the reversible nature of the covalent imine bonds formed between the aldehyde groups of OA and the amine groups present in the DNA nucleotides. Moreover, the incorporation of charged nSi further enhanced the shear strength of the formulated hydrogels by establishing electrostatic interactions with the phosphate groups of the DNA network. The optimized hydrogel was able to promote the sustained release of simvastatin for more than a week. The bioactivity of the released drug was confirmed by testing its ability to induce osteogenic differentiation and migration of human adipose-derived stem cells in vitro. Overall, the results obtained from this study demonstrate that DNA could be used as a natural biopolymer to fabricate self-healing injectable hydrogels with sustained release properties for minimally invasive therapeutic approaches. STATEMENT OF SIGNIFICANCE: Dynamic covalent chemistry, especially Schiff base reactions have emerged as a promising route for the formation of injectable hydrogels. Our study demonstrated the development of a DNA-based self-healing hydrogel formed via Schiff base reaction occurring at physiological conditions. The hydrogels functioned as sustained delivery vehicles for the hydrophobic drug simvastatin, which requires a polymeric carrier for controlled delivery of therapeutic concentrations of the drug without exhibiting cytotoxic effects. Presently available hydrogel-based drug delivery systems encounter major challenges for the delivery of hydrophobic drugs due to the hydrophilic nature of the base matrix. Our strategy presents a platform technology for the design of minimally invasive approaches for the sustained delivery of hydrophobic drugs similar to simvastatin.


Sujet(s)
ADN/composition chimique , Systèmes de délivrance de médicaments , Hydrogels/composition chimique , Injections , Alginates/composition chimique , Réactifs réticulants/composition chimique , Préparations à action retardée/pharmacologie , Libération de médicament , Humains , Imines/composition chimique , Nanoparticules/composition chimique , Oxydoréduction , Rhéologie , Silicates/composition chimique , Simvastatine/pharmacologie
14.
ACS Macro Lett ; 9(9): 1230-1236, 2020 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-35638638

RÉSUMÉ

This work investigates a sequential strategy to develop DNA-based hydrogel scaffolds with interpenetrating polymeric network. The scaffolds were formed via a two-step procedure. First, a covalently cross-linked DNA-based cryogel was formed by the chemical reaction between DNA strands and a bifunctional cross-linker, polyethylene glycol diepoxide at subzero temperatures. In the second step, alginate chains were absorbed into the preformed macroporous DNA cryogel network, followed by ionic cross-linking with divalent calcium ions. The individual and synergistic effects of covalent and ionic cross-linkings on mechanical and physical properties of the IPN cryogel were tested. The IPN cryogels were able to sustain large deformations higher than 95% of strain under compressive forces without exhibiting any failure. Addition of a physically cross-linked alginate network to the covalently linked DNA cryogel significantly enhanced its toughness and energy dissipation compared to the covalent network alone. The formulated hydrogels also exhibited excellent biocompatibility with human stem cells. Overall, this DNA-based IPN cryogel has the potential to be used as a biomaterial scaffold for a diverse range of tissue engineering applications.

15.
ACS Appl Bio Mater ; 3(2): 945-951, 2020 Feb 17.
Article de Anglais | MEDLINE | ID: mdl-35019296

RÉSUMÉ

Gellan gum-based hydrogels display limited cell adhesion ability due to the absence of cell-anchorage points usually present in proteins found in the extracellular matrix (ECM). This issue limits their use in the biomedical field as scaffolds to promote tissue repair. Our work addresses this challenge by investigating the use of polydopamine (pDA) as a bioactive layer to improve the surface and biological properties of gellan gum-based hydrogels cross-linked using carbodiimide chemistry. Upon treatment with a pDA layer, the hydrogel displayed an increase in wettability and swelling properties. This change in physical properties had a direct impact on the biological properties of the scaffolds. Precisely, human adipose-derived stem cells (hASCs) seeded on the pDA coated gellan gum hydrogels displayed larger cell area, increased proliferation rate, and enhanced gene expression of focal adhesion and cytoskeletal proteins. Overall, the findings of this research support the use of pDA coating as a possible approach to improve the biological features of gellan gum-based scaffolds and modulate stem cell morphology and proliferation.

16.
Cardiovasc Eng Technol ; 10(4): 628-637, 2019 12.
Article de Anglais | MEDLINE | ID: mdl-31650518

RÉSUMÉ

PURPOSE: Conventional methods of seeding decellularized heart valves for heart valve tissue engineering have led to inconsistent results in interstitial cellular repopulation, particularly of the distal valve leaflet, and notably distinct from documented re-endothelialization. The use of bioreactor conditioning mimicking physiologic parameters has been well explored but cellular infiltration remains challenging. Non-characteristic, non-physiologic conditioning parameters within a bioreactor, such as hypoxia and cyclic chamber pressure, may be used to increase the cellular infiltration leading to increased recellularization. METHODS: To investigate the effects of novel and perhaps non-intuitive bioreactor conditioning parameters, ovine aortic heart valves were seeded with mesenchymal stem cells and cultured in one of four environments: hypoxia and high cyclic pressures (120 mmHg), normoxia and high cyclic pressures, hypoxia and negative cyclic pressures (- 20 mmHg), and normoxia and negative cyclic pressures. Analysis included measurements of cellular density, cell phenotype, and biochemical concentrations. RESULTS: The results revealed that the bioreactor conditioning parameters influenced the degree of recellularization. Groups that implemented hypoxic conditioning exhibited increased cellular infiltration into the valve leaflet tissue compared to normoxic conditioning, while pressure conditioning did not have a significant effect of recellularization. Protein expression across all groups was similar, exhibiting a stem cell and valve interstitial cell phenotype. Biochemical analysis of the extracellular matrix was similar between all groups. CONCLUSION: These results suggest the use of non-physiologic bioreactor conditioning parameters can increase in vitro recellularization of tissue engineered heart valve leaflets. Particularly, hypoxic culture was found to increase the cellular infiltration. Therefore, bioreactor conditioning of tissue engineered constructs need not always mimic physiologic conditions, and it is worth investigating novel or uncharacteristic culture conditions as they may benefit aspects of tissue culture.


Sujet(s)
Valve aortique/physiologie , Bioprothèse , Bioréacteurs , Prothèse valvulaire cardiaque , Cellules souches mésenchymateuses/physiologie , Techniques de culture de tissus/instrumentation , Ingénierie tissulaire/instrumentation , Animaux , Valve aortique/cytologie , Hypoxie cellulaire , Cellules cultivées , Matrice extracellulaire/physiologie , Humains , Phénotype , Pression , Ovis aries
17.
ACS Appl Mater Interfaces ; 11(38): 34621-34633, 2019 Sep 25.
Article de Anglais | MEDLINE | ID: mdl-31483598

RÉSUMÉ

In situ tissue repair holds great potential as a cell-free regenerative strategy. A critical aspect of this approach is the selection of cell instructive materials that can efficiently regulate the defect microenvironment via the release of chemoattractant factors to mobilize and recruit endogenous stem cells toward the site of implantation. Here we report the design of a DNA-based hydrogel as a drug delivery platform for the sustained release of a promising chemoattractant, SDF-1α. The hydrogel is composed of chemically cross-linked DNA strands, which are bridged via silicate nanodisks (nSi). Silicate nanodisks electrostatically interact with the negatively charged DNA backbone resulting in the formation of a dual cross-linked nanocomposite hydrogel with a combination of chemical and physical cross-link points. The formulated nanocomposites display enhanced elasticity and mechanical toughness as compared to their nonsilicate containing counterparts. Moreover, the electrostatic interaction between nSi and SDF-1α leads to sustained release of the chemokine from the hydrogels. The in vitro bioactivity assays confirm the retention of chemotactic properties of the protein after its release. Overall, the dual cross-linked DNA-based hydrogel platform could be potentially used as a cell-instructive material for the recruitment of host stem cells to guide the process of in situ tissue repair.


Sujet(s)
Mouvement cellulaire/effets des médicaments et des substances chimiques , Chimiokine CXCL12 , ADN/composition chimique , Hydrogels/composition chimique , Nanostructures/composition chimique , Silicates/composition chimique , Cellules souches/métabolisme , Animaux , Chimiokine CXCL12/composition chimique , Chimiokine CXCL12/pharmacocinétique , Chimiokine CXCL12/pharmacologie , Humains , Souris , Cellules RAW 264.7 , Cellules souches/cytologie
18.
Mol Pharm ; 16(10): 4302-4312, 2019 10 07.
Article de Anglais | MEDLINE | ID: mdl-31398053

RÉSUMÉ

Secretome-based therapies have the potential to become the next generation of viable therapeutic wound repair treatments. However, precise strategies aimed to refine and control the secretome composition are necessary to enhance its therapeutic efficacy and facilitate clinical translation. In this study, we aim to accomplish this by transfecting human adipose-derived stem cells (hASCs) with microRNA-146a, which is a potent regulator of angiogenesis and inflammation. The secretome composition obtained from the transfected hASCs (secretome146a) was characterized and compared to nontransfected hASCs secretome to evaluate changes in angiogenic and anti-inflammatory growth factor, cytokine, and miRNA content. In vitro proliferation, migration, and tubular morphogenesis assays using human umbilical vein endothelial cells (HUVECs) were completed to monitor the proangiogenic efficacy of the secretome146a. Finally, the anti-inflammatory efficacy of the secretome146a was assessed using HUVECs that were activated to an inflammatory state by IL-1ß. The resulting HUVEC gene expression and protein activity of key inflammatory mediators were evaluated before and after secretome treatment. Overall, the secretome146a contained a greater array and concentration of therapeutic paracrine molecules, which translated into a superior angiogenic and anti-inflammatory efficacy. Therefore, this represents a promising strategy to produce therapeutic secretome for the promotion of wound repair processes.


Sujet(s)
Tissu adipeux/métabolisme , Cellules endothéliales de la veine ombilicale humaine/métabolisme , microARN/génétique , Néovascularisation physiologique , Cellules souches/métabolisme , Cicatrisation de plaie , Tissu adipeux/cytologie , Mouvement cellulaire , Prolifération cellulaire , Cellules cultivées , Cellules endothéliales de la veine ombilicale humaine/cytologie , Humains , Interleukine-1 bêta/génétique , Interleukine-1 bêta/métabolisme , microARN/métabolisme , Cellules souches/cytologie
19.
J Am Heart Assoc ; 8(15): e012089, 2019 08 06.
Article de Anglais | MEDLINE | ID: mdl-31315484

RÉSUMÉ

Background Myocardial infarction results in a large-scale cardiomyocyte loss and heart failure due to subsequent pathological remodeling. Whereas zebrafish and neonatal mice have evident cardiomyocyte expansion following injury, adult mammalian cardiomyocytes are principally nonproliferative. Despite historical presumptions of stem cell-mediated cardiac regeneration, numerous recent studies using advanced lineage-tracing methods demonstrated that the only source of cardiomyocyte renewal originates from the extant myocardium; thus, the augmented proliferation of preexisting adult cardiomyocytes remains a leading therapeutic approach toward cardiac regeneration. In the present study we investigate the significance of suppressing cell cycle inhibitors Rb1 and Meis2 to promote adult cardiomyocyte reentry to the cell cycle. Methods and Results In vitro experiments with small interfering RNA-mediated simultaneous knockdown of Rb1 and Meis2 in both adult rat cardiomyocytes, isolated from 12-week-old Fischer rats, and human induced pluripotent stem cell-derived cardiomyocytes showed a significant increase in cell number, a decrease in cell size, and an increase in mononucleated cardiomyocytes. In vivo, a hydrogel-based delivery method for small interfering RNA-mediated silencing of Rb1 and Meis2 is utilized following myocardial infarction. Immunofluorescent imaging analysis revealed a significant increase in proliferation markers 5-ethynyl-2'-deoxyuridine, PH3, KI67, and Aurora B in adult cardiomyocytes as well as improved cell survivability with the additional benefit of enhanced peri-infarct angiogenesis. Together, this intervention resulted in a reduced infarct size and improved cardiac function post-myocardial infarction. Conclusions Silencing of senescence-inducing pathways in adult cardiomyocytes via inhibition of Rb1 and Meis2 results in marked cardiomyocyte proliferation and increased protection of cardiac function in the setting of ischemic injury.


Sujet(s)
Cycle cellulaire/physiologie , Protéines à homéodomaine/génétique , Infarctus du myocarde , Myocytes cardiaques/cytologie , Protéines de liaison à la protéine du rétinoblastome/génétique , Facteurs de transcription/génétique , Ubiquitin-protein ligases/génétique , Facteurs âges , Animaux , Protéines à homéodomaine/physiologie , Humains , Mâle , Infarctus du myocarde/anatomopathologie , Infarctus du myocarde/physiopathologie , Rats , Rats de lignée F344 , Protéines de liaison à la protéine du rétinoblastome/physiologie , Facteurs de transcription/physiologie , Ubiquitin-protein ligases/physiologie
20.
Cell Mol Bioeng ; 11(3): 211-217, 2018 Jun.
Article de Anglais | MEDLINE | ID: mdl-30338007

RÉSUMÉ

INTRODUCTION: Physical and mechanical properties of ceramic-based scaffolds can be modulated by introducing hydrogel coatings on their surface. For instance, hydrogels can be used as elastic layers to overcome the brittleness of synthetic ceramic materials or to control the delivery of essential osteogenic factors. In this work, we aimed to achieve both goals by fabricating a novel cytocompatible hydrogel made of gelatin-alginate as a coating for beta-tricalcium phosphate (ß-TCP) scaffolds. METHODS: The hydrogel synthesis was optimized by varying the concentration of the crosslinkers N-hydroxysuccinimide and N-Ethyl-N'-(3-dimethyl aminopropyl) carbodiimide (NHS/EDC). Swelling, degradability and mechanical studies were carried out to identify the suitable hydrogel coating formulation for the ß-TCP scaffolds. The cytocompatibility of the coated ceramic was assessed in vitro by testing the proliferation and the osteogenic differentiation of human adipose stem cell (hASCs) for two weeks. RESULTS: The designed hydrogel layer could withstand cyclic compression and protected the brittle internal core of the ceramic. The hydrogel coating modulated the diffusion of the model protein BSA according to the degree of crosslinking of the hydrogel layer. Additionally, the polymeric network was able to retain positively charged proteins such as lysozyme due to the strong electrostatic interactions with carboxylic groups of alginate. A higher expression of alkaline phosphates activity was found on hASCs seeded on the coated scaffolds compared to the hydrogels without any ß-TCP. CONCLUSION: Overall, the hydrogel coating characterized in this study represents a valid strategy to overcome limitations of brittle ceramic-based materials used as scaffolds for bone tissue engineering applications.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...