Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Bioact Mater ; 38: 540-558, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38872731

ABSTRACT

Bacteria can be programmed to deliver natural materials with defined biological and mechanical properties for controlling cell growth and differentiation. Here, we present an elastic, resilient and bioactive polysaccharide derived from the extracellular matrix of Pantoea sp. BCCS 001. Specifically, it was methacrylated to generate a new photo crosslinkable hydrogel that we coined Pantoan Methacrylate or put simply PAMA. We have used it for the first time as a tissue engineering hydrogel to treat VML injuries in rats. The crosslinked PAMA hydrogel was super elastic with a recovery nearing 100 %, while mimicking the mechanical stiffness of native muscle. After inclusion of thiolated gelatin via a Michaelis reaction with acrylate groups on PAMA we could also guide muscle progenitor cells into fused and aligned tubes - something reminiscent of mature muscle cells. These results were complemented by sarcomeric alpha-actinin immunostaining studies. Importantly, the implanted hydrogels exhibited almost 2-fold more muscle formation and 50 % less fibrous tissue formation compared to untreated rat groups. In vivo inflammation and toxicity assays likewise gave rise to positive results confirming the biocompatibility of this new biomaterial system. Overall, our results demonstrate that programmable polysaccharides derived from bacteria can be used to further advance the field of tissue engineering. In greater detail, they could in the foreseeable future be used in practical therapies against VML.

2.
Adv Biol (Weinh) ; 8(6): e2300621, 2024 06.
Article in English | MEDLINE | ID: mdl-38580620

ABSTRACT

Hyaluronic acid (HA) plays a prominent role in various aspects of reproductive biology and assisted reproductive technologies (ART). This review describes the multifaceted influence of HA, ranging from primordial germ cell migration, ovarian follicle development, and ovulation in females to sperm structure, physiology, motility, and capacitation in males. In addition, HA also plays an important role in fertilization and promotes embryo implantation by mediating cellular adhesion and communication within the uterus. Against this physiological background, the review examines the current applications of HA in the context of ART. In addition, the article addresses the emerging field of reproductive tissue engineering, where HA-based hydrogels offer promising perspectives as they can support the development of mature oocytes and spermatogenesis in vitro. Overall, this review highlights the integral role of HA in the intricate mechanisms of reproductive biology and its growing importance for improving ART outcomes and the field of tissue engineering of the reproductive system.


Subject(s)
Hyaluronic Acid , Reproductive Techniques, Assisted , Tissue Engineering , Hyaluronic Acid/chemistry , Hyaluronic Acid/metabolism , Humans , Tissue Engineering/methods , Female , Male , Animals , Reproduction/physiology
3.
ACS Biomater Sci Eng ; 10(2): 800-813, 2024 02 12.
Article in English | MEDLINE | ID: mdl-38159039

ABSTRACT

Light-cured conductive hydrogels have attracted immense interest in the regeneration of electroactive tissues and bioelectronic interfaces. Despite the unique properties of MXene (MX), its light-blocking effect in the range of 300-600 nm hinders the efficient cross-linking of photocurable hydrogels. In this study, we investigated the photo-cross-linking process of MX-gelatin methacrylate (GelMa) composites with different types of photoinitiators and MX concentrations to prepare biocompatible, injectable, conductive, and photocurable composite hydrogels. The examined photoinitiators were Eosin Y, Irgacure 2959 (Type I), and lithium phenyl-2,4,6-trimethylbenzoyl phosphinate (Type II). The light-blocking effect of MX strongly affected the thickness, pore structure, swelling ratio, degradation, and mechanical properties of the light-cured hydrogels. Uniform distribution of MX in the hydrogel matrix was achieved at concentrations up to 0.04 wt % but the film thickness and curing times varied depending on the type of photoinitiator. It was feasible to prepare thin films (0.5 mm) by employing Type I photoinitiators under a relatively long light irradiation (4-5 min) while thick films with centimeter sizes could be rapidly cured by using Type II photoinitiator (<60 s). The mechanical properties, including elastic modulus, toughness, and stress to break for the Type II hydrogels were significantly superior (up to 300%) to those of Type I hydrogels depending on the MX concentration. The swelling ratio was also remarkably higher (648-1274%). A conductivity of about 1 mS/cm was attained at 0.1 mg/mL MX for the composite hydrogel cured by the Type I photoinitiator. In vitro cytocompatibility assays determined that the hydrogels promoted cell viability, metabolic activity, and robust proliferation of C2C12 myoblasts, which indicated their potential to support muscle cell growth during myogenesis. The developed photocurable GelMa-MX hydrogels have the potential to serve as bioactive and conductive scaffolds to modulate cellular functions and for tissue-device interfacing.


Subject(s)
Biocompatible Materials , Hydrogels , Nitrites , Transition Elements , Biocompatible Materials/pharmacology , Hydrogels/pharmacology , Hydrogels/chemistry , Electric Conductivity , Cell Survival , Gelatin/chemistry , Methacrylates/chemistry , Methacrylates/pharmacology
4.
Bioengineering (Basel) ; 10(12)2023 Dec 18.
Article in English | MEDLINE | ID: mdl-38136029

ABSTRACT

Among the various biochemical and biophysical inducers for neural regeneration, electrical stimulation (ES) has recently attracted considerable attention as an efficient means to induce neuronal differentiation in tissue engineering approaches. The aim of this in vitro study was to develop a nanofibrous scaffold that enables ES-mediated neuronal differentiation in the absence of exogenous soluble inducers. A nanofibrous scaffold composed of polycaprolactone (PCL), poly-L-lactic acid (PLLA), and single-walled nanotubes (SWNTs) was fabricated via electrospinning and its physicochemical properties were investigated. The cytocompatibility of the electrospun composite with the PC12 cell line and bone marrow-derived mesenchymal stem cells (BMSCs) was investigated. The results showed that the PCL/PLLA/SWNT nanofibrous scaffold did not exhibit cytotoxicity and supported cell attachment, spreading, and proliferation. ES was applied to cells cultured on the nanofibrous scaffolds at different intensities and the expression of the three neural markers (Nestin, Microtubule-associated protein 2, and ß tubulin-3) was evaluated using RT-qPCR analysis. The results showed that the highest expression of neural markers could be achieved at an electric field intensity of 200 mV/cm, suggesting that the scaffold in combination with ES can be an efficient tool to accelerate neural differentiation in the absence of exogenous soluble inducers. This has important implications for the regeneration of nerve injuries and may provide insights for further investigations of the mechanisms underlying ES-mediated neuronal commitment.

5.
Exp Cell Res ; 431(2): 113766, 2023 10 15.
Article in English | MEDLINE | ID: mdl-37678504

ABSTRACT

Stem cells in their natural microenvironment are exposed to biochemical and biophysical cues emerging from the extracellular matrix (ECM) and neighboring cells. In particular, biomechanical forces modulate stem cell behavior, biological fate, and early developmental processes by sensing, interpreting, and responding through a series of biological processes known as mechanotransduction. Local structural changes in the ECM and mechanics are driven by reciprocal activation of the cell and the ECM itself, as the initial deposition of matrix proteins sequentially affects neighboring cells. Recent studies on stem cell mechanoregulation have provided insight into the importance of biomechanical signals on proper tissue regeneration and function and have shown that precise spatiotemporal control of these signals exists in stem cell niches. Against this background, the aim of this work is to review the current understanding of the molecular basis of mechanotransduction by analyzing how biomechanical forces are converted into biological responses via cellular signaling pathways. In addition, this work provides an overview of advanced strategies using stem cells and biomaterial scaffolds that enable precise spatial and temporal control of mechanical signals and offer great potential for the fields of tissue engineering and regenerative medicine will be presented.


Subject(s)
Cues , Tissue Engineering , Mechanotransduction, Cellular , Regenerative Medicine , Stem Cells
6.
Biotechnol Bioeng ; 120(11): 3396-3408, 2023 11.
Article in English | MEDLINE | ID: mdl-37526327

ABSTRACT

During normal urination, smooth muscle cells (SMCs) in the lower urinary tract (LUT) are exposed to mechanical signals that have a critical impact on tissue structure and function. Nevertheless, the mechanisms underlying the maintenance of the contractile phenotype of SMCs remain poorly understood. This is due, in part, to a lack of studies that have examined the effects of mechanical loading using three-dimensional (3D) models. In this study, surface modifications of polydimethylsiloxane (PDMS) membrane were evaluated to investigate the effects of cyclic mechanical stimulation on SMC maturation in 3D constructs. Commercially available cell stretching plates were modified with amino or methacrylate groups to promote adhesion of 3D constructs fabricated by bioprinting. After 6 days of stimulation, the effects of mechanical stimulation on the expression of contractile markers at the mRNA and protein levels were analyzed. Methacrylate-modified surfaces supported stable adhesion of the 3D constructs to the membrane and facilitated cyclic mechanical stimulation, which significantly increased the expression of contractile markers at the mRNA and protein levels. These effects were found to be mediated by activation of the p38 MAPK pathway, as inhibition of this pathway abolished the effects of stimulation in a dose-dependent manner. These results provide valuable insights into the role of mechanical signaling in maintaining the contractile phenotype of bladder SMCs, which has important implications for the development of future treatments for LUT diseases.


Subject(s)
Bioprinting , Hydrogels , Hydrogels/chemistry , Muscle, Smooth , Myocytes, Smooth Muscle , Dimethylpolysiloxanes/pharmacology , Methacrylates/pharmacology , RNA, Messenger , Tissue Engineering/methods , Bioprinting/methods , Printing, Three-Dimensional , Tissue Scaffolds/chemistry
7.
Bioengineering (Basel) ; 10(5)2023 May 09.
Article in English | MEDLINE | ID: mdl-37237636

ABSTRACT

Excessive skin scarring affects over 100 million patients worldwide, with effects ranging from cosmetic to systemic problems, and an effective treatment is yet to be found. Ultrasound-based therapies have been used to treat a variety of skin disorders, but the exact mechanisms behind the observed effects are still unclear. The aim of this work was to demonstrate the potential of ultrasound for the treatment of abnormal scarring by developing a multi-well device based on printable piezoelectric material (PiezoPaint™). First, compatibility with cell cultures was evaluated using measurements of heat shock response and cell viability. Second, the multi-well device was used to treat human fibroblasts with ultrasound and quantify their proliferation, focal adhesions, and extracellular matrix (ECM) production. Ultrasound caused a significant reduction in fibroblast growth and ECM deposition without changes in cell viability or adhesion. The data suggest that these effects were mediated by nonthermal mechanisms. Interestingly, the overall results suggest that ultrasound treatment would a be beneficial therapy for scar reduction. In addition, it is expected that this device will be a useful tool for mapping the effects of ultrasound treatment on cultured cells.

9.
Bioeng Transl Med ; 8(2): e10383, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36925674

ABSTRACT

Tissue engineering (TE) is currently considered a cutting-edge discipline that offers the potential for developing treatments for health conditions that negatively affect the quality of life. This interdisciplinary field typically involves the combination of cells, scaffolds, and appropriate induction factors for the regeneration and repair of damaged tissue. Cell fate decisions, such as survival, proliferation, or differentiation, critically depend on various biochemical and biophysical factors provided by the extracellular environment during developmental, physiological, and pathological processes. Therefore, understanding the mechanisms of action of these factors is critical to accurately mimic the complex architecture of the extracellular environment of living tissues and improve the efficiency of TE approaches. In this review, we recapitulate the effects that biochemical and biophysical induction factors have on various aspects of cell fate. While the role of biochemical factors, such as growth factors, small molecules, extracellular matrix (ECM) components, and cytokines, has been extensively studied in the context of TE applications, it is only recently that we have begun to understand the effects of biophysical signals such as surface topography, mechanical, and electrical signals. These biophysical cues could provide a more robust set of stimuli to manipulate cell signaling pathways during the formation of the engineered tissue. Furthermore, the simultaneous application of different types of signals appears to elicit synergistic responses that are likely to improve functional outcomes, which could help translate results into successful clinical therapies in the future.

10.
Tissue Eng Regen Med ; 20(2): 199-212, 2023 04.
Article in English | MEDLINE | ID: mdl-36401768

ABSTRACT

BACKGROUND: Reliable in vitro cellular models are needed to study the phenotypic modulation of smooth muscle cells (SMCs) in health and disease. The aim of this study was to optimize gelatin methacrylate (GelMA)/alginate hydrogels for bioprinting three-dimensional (3D) SMC constructs. METHODS: Four different hydrogel groups were prepared by mixing different concentrations (% w/v) of GelMA and alginate: G1 (5/1.5), G2 (5/3), G3 (7.5/1.5), and G4 (7.5/3). GelMA 10% was used as control (G5). A circular structure containing human bladder SMCs was fabricated by using an extrusion-based bioprinter. The effects of the mixing ratios on printability, viability, proliferation, and differentiation of the cells were investigated. RESULTS: Rheological analysis showed that the addition of alginate significantly stabilized the change in mechanical properties with temperature variations. The group with the highest GelMA and alginate concentrations (G4) exhibited the highest viscosity, resulting in better stability of the 3D construct after crosslinking. Compared to other hydrogel compositions, cells in G4 maintained high viability (> 80%), exhibited spindle-shaped morphology, and showed a significantly higher proliferation rate within an 8-day period. More importantly, G4 provided an optimal environment for the induction of a SMC contractile phenotype, as evidenced by significant changes in the expression of marker proteins and morphological parameters. CONCLUSION: Adjusting the composition of GelMA/alginate hydrogels is an effective means of controlling the SMC phenotype. These hydrogels support bioprinting of 3D models to study phenotypic smooth muscle adaptation, with the prospect of using the constructs in the study of therapies for the treatment of urethral strictures.


Subject(s)
Bioprinting , Hydrogels , Humans , Hydrogels/chemistry , Cell Differentiation , Bioprinting/methods , Gelatin/chemistry , Alginates/chemistry , Methacrylates/pharmacology , Methacrylates/chemistry , Muscle, Smooth
11.
Int J Mol Sci ; 23(22)2022 Nov 15.
Article in English | MEDLINE | ID: mdl-36430557

ABSTRACT

Urethral stricture is a common urinary tract disorder in men that can be caused by iatrogenic causes, trauma, inflammation, or infection and often requires reconstructive surgery. The current therapeutic approach for complex urethral strictures usually involves reconstruction with autologous tissue from the oral mucosa. With the goal of overcoming the lack of sufficient autologous tissue and donor site morbidity, research over the past two decades has focused on cell-based tissue-engineered substitutes. While the main focus has been on autologous cells from the penile tissue, bladder, and oral cavity, stem cells from sources such as adipose tissue and urine are competing candidates for future urethral regeneration due to their ease of collection, high proliferative capacity, maturation potential, and paracrine function. This review addresses the sources, advantages, and limitations of cells for tissue engineering in the urethra and discusses recent approaches to improve cell survival, growth, and differentiation by mimicking the mechanical and biophysical properties of the extracellular environment.


Subject(s)
Tissue Engineering , Urethra , Male , Humans , Conditioning, Psychological , Urinary Bladder , Penis
12.
EMBO J ; 41(17): e111650, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35899396

ABSTRACT

Mechanical inputs give rise to p38 and JNK activation, which mediate adaptive physiological responses in various tissues. In skeletal muscle, contraction-induced p38 and JNK signaling ensure adaptation to exercise, muscle repair, and hypertrophy. However, the mechanisms by which muscle fibers sense mechanical load to activate this signaling have remained elusive. Here, we show that the upstream MAP3K ZAKß is activated by cellular compression induced by osmotic shock and cyclic compression in vitro, and muscle contraction in vivo. This function relies on ZAKß's ability to recognize stress fibers in cells and Z-discs in muscle fibers when mechanically perturbed. Consequently, ZAK-deficient mice present with skeletal muscle defects characterized by fibers with centralized nuclei and progressive adaptation towards a slower myosin profile. Our results highlight how cells in general respond to mechanical compressive load and how mechanical forces generated during muscle contraction are translated into MAP kinase signaling.


Subject(s)
Mitogen-Activated Protein Kinases , Muscle, Skeletal , Animals , MAP Kinase Kinase Kinases , Mice , Mitogen-Activated Protein Kinases/metabolism , Muscle Contraction/physiology , Muscle, Skeletal/metabolism , Phosphorylation , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/genetics
13.
Cells ; 11(7)2022 04 06.
Article in English | MEDLINE | ID: mdl-35406800

ABSTRACT

It has been suggested that immunophenotypically defined lineages within the in vitro expanded adipose-derived stem cell (ASC) may play a beneficial role from the perspective of a personalized intervention. Therefore, to better understand the implications of different surface marker profiles for the functionality, we set out to examine the evolution of ASC-variants based on the co-expression of five bright or eight dim epitopes. At passages P1, P4, and P8, the co-localization of five bright markers (CD73, CD90, CD105, CD166, and CD201), or eight dim markers (CD34, CD36, CD200, CD248, CD271, CD274, CD146, and the Stro-1), was investigated by flow cytometry. Selected subpopulations were isolated using the fluorescence-activated cells sorting from the cryopreserved P4 and analyzed in terms of proliferative and clonogenic properties, trilineage differentiation, and wound healing potential. Only two of the dim epitopes were found in representative subpopulations (SP), and from the P4 onwards, two major combinations featuring the CD274+ (SP1) or the CD274+ CD146+ (SP2) emerged. Upon sorting and growth, both subpopulations assumed new but highly similar clonal profiles, consisting of the CD274+ CD146+ and the CD274+ CD146+ CD248+ phenotypes. The functional analysis revealed that the SP2 surpassed SP1 and the unfractionated cells regarding the growth rate, clonogenic activity, and the wound closure and endothelial tube formation potential. The surface epitopes may be considered a tool to enrich specific functionality and thus improve therapeutic outcomes in dedicated circumstances.


Subject(s)
Adipose Tissue , Stem Cells , Adipose Tissue/metabolism , Biomarkers/metabolism , CD146 Antigen/metabolism , Epitopes/metabolism , Wound Healing
14.
Physiol Rep ; 9(21): e15077, 2021 11.
Article in English | MEDLINE | ID: mdl-34713978

ABSTRACT

Insight into the bidirectional signaling between primary human myogenic cells and neurons is lacking. For this purpose, human myogenic cells were derived from the semitendinosus and gracilis muscles of five healthy individuals and co-cultured with cerebellar granule neurons from two litters of 7-day-old Wistar rat pups, in muscle medium or neural medium, alongside monocultures of myogenic cells or neurons. RT-PCR was performed to determine human mRNA levels of GAPDH, Ki67, myogenin, and MUSK, and the acetylcholine receptor subtypes CHRNA1, CHRNB1, CHRNG, CHRND, and CHRNE, and rat mRNA levels of GAPDH, Fth1, Rack1, vimentin, Cdh13, and Ppp1r1a. Immunocytochemistry was used to evaluate neurite outgrowth (GAP43) in the presence and absence of myogenic cells. Co-culture with primary neurons lead to higher myogenic cell gene expression levels of GAPDH, myogenin, MUSK, CHRNA1, CHRNG, and CHRND, compared to myogenic cells cultured alone. It appeared that neurons preferentially attached to myotubes and that neurite outgrowth was enhanced when neurons were cultured with myogenic cells compared to monoculture. In neural medium, rat mRNA levels of GAPDH, vimentin, Cdh13, and Ppp1r1a were greater in co-culture, versus monoculture, whereas in muscle medium co-culture lead to lower levels of Fth1, Rack1, vimentin, and Cdh13 than monoculture. These findings demonstrate mutually beneficial stimulatory signaling between rat cerebellar granule neurons and human myogenic cells, providing support for an active role for both the neuron and the muscle cell in stimulating neurite growth and myogenesis. Bidirectional muscle nerve signaling.


Subject(s)
Cell Communication , Myoblasts/metabolism , Neurons/cytology , Neurons/metabolism , Signal Transduction , Adolescent , Adult , Animals , Cells, Cultured , Cerebellum/cytology , Coculture Techniques/methods , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/genetics , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Humans , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Middle Aged , Myoblasts/cytology , Myogenin/genetics , Myogenin/metabolism , Neuronal Outgrowth , Rats , Rats, Wistar , Receptors, Cholinergic/genetics , Receptors, Cholinergic/metabolism , Vimentin/genetics , Vimentin/metabolism
15.
Animals (Basel) ; 11(8)2021 Aug 21.
Article in English | MEDLINE | ID: mdl-34438913

ABSTRACT

Preclinical research within the area of urethral tissue engineering has not yet been successfully translated into an efficient therapeutic option for patients. This gap could be attributed, in part, to inadequate design and reporting of the studies employing laboratory animals. In this study, a systematic review was conducted to investigate the quality of reporting in preclinical studies utilizing tissue engineering approaches for urethral repair. The scope was on studies performed in rabbits, published between January 2014 and March 2020. Quality assessment of the data was conducted according to the Animal Research: Reporting of in Vivo Experiments (ARRIVE) guidelines by the scoring of a 38-item checklist in different categories. A total of 28 articles that fulfilled the eligibility criteria were included in the study. The range of ARRIVE score was from 0 to 100, taking into consideration having reported the item in question or not. The mean checklist score was 53%. The items that attained the highest scores included the number of animals utilized, the size of control and experimental groups, and the definition of experimental outcomes. The least frequently reported items included the data regarding the experimental procedure, housing and husbandry, determination and justification of the number of animals, and reporting of adverse events. Surprisingly, full disclosure about ethical guidelines and animal protocol approval was missing in 54% of the studies. No paper stated the sample size estimation. Overall, our study found that a large number of studies display inadequate reporting of fundamental information and that the quality of reporting improved marginally over the study period. We encourage a comprehensive implementation of the ARRIVE guidelines in animal studies exploring tissue engineering for urethral repair, not only to facilitate effective translation of preclinical research findings into clinical therapies, but also to ensure compliance with ethical principles and to minimize unnecessary animal studies.

16.
Cells ; 10(2)2021 01 22.
Article in English | MEDLINE | ID: mdl-33499095

ABSTRACT

In order to enhance the therapeutic potential, it is important that sufficient knowledge regarding the dynamic changes of adipose-derived stem cell (ASC) immunophenotypical and biological properties during in vitro growth is available. Consequently, we embarked on a study to follow the evolution of highly defined cell subsets from three unrelated donors in the course of eight passages on tissue culture polystyrene. The co-expression patterns were defined by panels encompassing seven and five cell surface markers, including CD34, CD146, CD166, CD200, CD248, CD271, and CD274 and CD29, CD31, CD36, CD201, and Stro-1, respectively. The analysis was performed using multichromatic flow cytometry. We observed a major paradigm shift, where the CD166-CD34+ combination which was found across all cell subsets early in the culture was replaced by the CD166+ phenotype as the population homogeneity increased with time. At all analysis points, the cultures were dominated by a few major clones that were highly prevalent in most of the donors. The selection process resulted in two predominant clones in the larger panel (CD166+CD34-CD146-CD271- CD274-CD248-CD200- and CD166+CD34+ CD146-CD271-CD274-CD248-CD200-) and one clone in the smaller panel (CD29+CD201+CD36- Stro-1- CD31-). The minor subsets, including CD166+CD34-CD146-CD271+CD274-CD248-CD200- and CD166+CD34+CD146+CD271-CD274-CD248-CD200-, and CD29+CD201-CD36-Stro-1-CD31-, CD29+CD201+CD36-Stro-1+CD31-, and CD29+CD201+CD36+Stro-1-CD31-, in the seven and five marker panels, respectively, were, on the other, hand highly fluctuating and donor-dependent. The results demonstrate that only a limited number of phenotypical repertoires are possible in ASC cultures. Marked differences in their relative occurrence between distinct individuals underscore the need for potency standardization of different ASC preparation to improve the clinical outcome.


Subject(s)
Adipose Tissue/cytology , Immunophenotyping , Stem Cells/cytology , Biomarkers/metabolism , Cell Proliferation , Cells, Cultured , Humans , Tissue Donors
17.
Methods ; 171: 68-76, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31299290

ABSTRACT

Chronic non-healing wounds are detrimental for the quality of life of the affected individuals and represent a major burden for the health care systems. Adipose-derived stem cells (ASCs) are being investigated for the development of novel treatments of chronic wounds, as they have shown several positive effects on wound healing. While these effects appear to be mediated by the release of soluble factors, it is has also become apparent that the extracellular matrix (ECM) deposited by ASCs is essential in several phases of the wound healing process. In this work, we describe an approach to produce ECM scaffolds derived from ASCs in culture. Upon growth of ASCs into an overconfluent cell layer, a detergent-based cell extraction approach is applied to remove the cellular components. The extraction is followed by an enzymatic treatment to remove the residual DNA. The resultant cell-derived scaffolds are depleted of cellular components, display low DNA remnant, and retain the native fibrillar organization of the ECM. Analysis of the molecular composition of the ECM scaffolds revealed that they are composed of collagens type I and III, and fibronectin. The decellularized scaffolds represent a substrate that supports adhesion and proliferation of primary human fibroblasts and dermal microvascular endothelial cells, indicating their potential as platforms for wound healing studies.


Subject(s)
Mesenchymal Stem Cells/cytology , Tissue Engineering , Tissue Scaffolds/chemistry , Wound Healing/drug effects , Adipocytes/cytology , Adipose Tissue/cytology , Adipose Tissue/transplantation , Animals , Endothelial Cells/cytology , Endothelial Cells/transplantation , Extracellular Matrix/chemistry , Extracellular Matrix/transplantation , Fibroblasts/drug effects , Fibronectins/chemistry , Humans , Mesenchymal Stem Cells/chemistry , Quality of Life
18.
Adv Sci (Weinh) ; 6(16): 1801664, 2019 Aug 21.
Article in English | MEDLINE | ID: mdl-31453048

ABSTRACT

Given their durability and long-term stability, self-healable hydrogels have, in the past few years, emerged as promising replacements for the many brittle hydrogels currently being used in preclinical or clinical trials. To this end, the incompatibility between hydrogel toughness and rapid self-healing remains unaddressed, and therefore most of the self-healable hydrogels still face serious challenges within the dynamic and mechanically demanding environment of human organs/tissues. Furthermore, depending on the target tissue, the self-healing hydrogels must comply with a wide range of properties including electrical, biological, and mechanical. Notably, the incorporation of nanomaterials into double-network hydrogels is showing great promise as a feasible way to generate self-healable hydrogels with the above-mentioned attributes. Here, the recent progress in the development of multifunctional and self-healable hydrogels for various tissue engineering applications is discussed in detail. Their potential applications within the rapidly expanding areas of bioelectronic hydrogels, cyborganics, and soft robotics are further highlighted.

19.
ACS Appl Mater Interfaces ; 11(13): 12283-12297, 2019 Apr 03.
Article in English | MEDLINE | ID: mdl-30864429

ABSTRACT

The emergence of nontoxic, eco-friendly, and biocompatible polymers derived from natural sources has added a new and exciting dimension to the development of low-cost and scalable biomaterials for tissue engineering applications. Here, we have developed a mechanically strong and durable hydrogel composed of an eco-friendly biopolymer that exists within the cell walls of fruits and plants. Its trade name is pectin, and it bears many similarities with natural polysaccharides in the native extracellular matrix. Specifically, we have employed a new pathway to transform pectin into a ultraviolet (UV)-cross-linkable pectin methacrylate (PEMA) polymer. To endow this hydrogel matrix with cell differentiation and cell spreading properties, we have also incorporated thiolated gelatin into the system. Notably, we were able to fine-tune the compressive modulus of this hydrogel in the range ∼0.5 to ∼24 kPa: advantageously, our results demonstrated that the hydrogels can support growth and viability for a wide range of three-dimensionally (3D) encapsulated cells that include muscle progenitor (C2C12), neural progenitor (PC12), and human mesenchymal stem cells (hMSCs). Our results also indicate that PEMA-gelatin-encapsulated hMSCs can facilitate the formation of bonelike apatite after 5 weeks in culture. Finally, we have demonstrated that PEMA-gelatin can yield micropatterned cell-laden 3D constructs through UV light-assisted lithography. The simplicity, scalability, processability, tunability, bioactivity, and low-cost features of this new hydrogel system highlight its potential as a stem cell carrier that is capable of bridging the gap between clinic and laboratory.


Subject(s)
Biocompatible Materials , Cells, Immobilized , Gelatin , Hydrogels , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells , Methacrylates , Pectins , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Differentiation/drug effects , Cell Line , Cells, Immobilized/cytology , Cells, Immobilized/metabolism , Cells, Immobilized/transplantation , Extracellular Matrix/chemistry , Gelatin/chemistry , Gelatin/pharmacology , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Methacrylates/chemistry , Methacrylates/pharmacology , PC12 Cells , Pectins/chemistry , Pectins/pharmacology , Rats
20.
Adv Exp Med Biol ; 1119: 1-19, 2018.
Article in English | MEDLINE | ID: mdl-30406362

ABSTRACT

Biomaterials are key components in tissue engineering and regenerative medicine applications, with the intended purpose of reducing the burden of disease and enhancing the quality of life of a large number of patients. The success of many regenerative medicine strategies, such as cell-based therapies, artificial organs, and engineered living tissues, is highly dependent on the ability to design or produce suitable biomaterials that can support and guide cells during tissue healing and remodelling processes. This chapter presents an overview about basic research concerning the use of different biomaterials for tissue engineering and regenerative medicine applications. Starting from a historical perspective, the chapter introduces the basic principles of designing biomaterials for tissue regeneration approaches. The main focus is set on describing the main classes of biomaterials that have been applied in regenerative medicine, including natural and synthetic polymers, bioactive ceramics, and composites. For each class of biomaterials, some of the most important physicochemical and biological properties are presented. Finally, some challenges and concerns that remain in this field are presented and discussed.


Subject(s)
Regenerative Medicine , Biocompatible Materials , Ceramics , Humans , Quality of Life , Tissue Engineering
SELECTION OF CITATIONS
SEARCH DETAIL
...