ArticleLiterature Review

Biomaterials for Recruiting and Activating Endogenous Stem Cells in situ Tissue Regeneration

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract

Over the past two decades in situ tissue engineering has emerged as a new approach where biomaterials are used to harness the body's own stem/progenitor cells to regenerate diseased or injured tissue. Immunomodulatory biomaterials are designed to promote a regenerative environment, recruit resident stem cells to diseased or injured tissue sites, and direct them towards tissue regeneration. This review explores advances gathered from in vitro and in vivo studies on in situ tissue regenerative therapies. Here we also examine the different ways this approach has been incorporated into biomaterial sciences in order to create customized biomaterial products for therapeutic applications in a broad spectrum of tissues and diseases. Statement of Significance Biomaterials can be designed to recruit stem cells and coordinate their behavior and function towards the restoration or replacement of damaged or diseased tissues in a process known as in situ tissue regeneration. Advanced biomaterial constructs with precise structure, composition, mechanical, and physical properties can be transplanted to tissue site and exploit local stem cells and their micro-environment to promote tissue regeneration. In the absence of cells, we explore the critical immunomodulatory, chemical and physical properties to consider in material design and choice. The application of biomaterials for in situ tissue regeneration has the potential to address a broad range of injuries and diseases.

No full-text available

Request Full-text Paper PDF

To read the full-text of this research,
you can request a copy directly from the authors.

... However, the ability of in vitro approach to reconstruct in vivo SwG microenvironment is limited. Due to the difficulty of in vitro engineering to maintain cell viability or phenotype, the potential for immune rejection and tumorigenesis [14], in vivo tissue regeneration is becoming increasingly attractive, which takes use of the body's natural capacity for tissue regeneration [15,16]. This process is aided by the biphasic calcium phosphate Ap-PLGA apatite-coated poly (lactic-co-glycolic acid) PLG poly (lactic-co-glycolic) acid pSmad (phosphorylated small mother against decapentaplegic) P(LLA-CL) poly(L-lactide-co-caprolactone) Fn fibronectin DFO des-ferrioxamine ADSCs adipose-derived mesenchymal stem cells PEG poly (ethylene glycol) CNCs cellulose nanocrystals EMs exosomes HUMSCs human umbilical cord mesenchymal stem cells exploitation of bioactive materials that can recruit host stem/progenitor cells to the wound site to guide the structural and functional restoration of injured tissues [17]. ...
... Homing factors are molecular factors that recruit stem cells to the wound site during stem cell homing. Stem cells can respond to gradients of chemo-attractants to be recruited to distant sites under the influence of homing factors to participate in wound healing [14,56]. Therefore, epidermal stem cells (EPSCs), myoepithelial stem cells, hair follicle stem cells, bone marrow mesenchymal stem cells (BM-MSCs) and other stem cells can potentially be recruited to the wound site to participate in SwG regeneration [47,49,50,57,58]. ...
... SwGs can be regenerated by stem cells, such as mesenchymal stem cells (MSCs), that can perceive mechanical cues from the microenvironment to promote cytoskeletal re-arrangement. This shows the potential to regulate the differentiation of stem cells via mechanotransduction and thus promote SwG regeneration [14]. Therefore, the stiffness and elasticity of bioactive materials should be well designed to modulate the behavior of cells with the potential to regenerate SwGs and guide their fate determination to promote SwG regeneration. ...
Article
Full-text available
Loss of sweat glands (SwGs) commonly associated with extensive skin defects is a leading cause of hyperthermia and heat stroke. In vivo tissue engineering possesses the potential to take use of the body natural ability to regenerate SwGs, making it more conducive to clinical translation. Despite recent advances in regenerative medicine, reconstructing SwG tissue with the same structure and function as native tissue remains challenging. Elucidating the SwG generation mechanism and developing biomaterials for in vivo tissue engineering is essential for understanding and developing in vivo SwG regenerative strategies. Here, we outline the cell biology associated with functional wound healing and the characteristics of bioactive materials. We critically summarize the recent progress in bioactive material-based cell modulation approaches for in vivo SwG regeneration, including the recruitment of endogenous cells to the skin lesion for SwG regeneration and in vivo cellular reprogramming for SwG regeneration. We discussed the re-establishment of microenvironment via bioactive material-mediated regulators. Besides, we offer promising perspectives for directing in situ SwG regeneration via bioactive material-based cell-free strategy, which is a simple and effective approach to regenerate SwG tissue with both fidelity of structure and function. Finally, we discuss the opportunities and challenges of in vivo SwG regeneration in detail. The molecular mechanisms and cell fate modulation of in vivo SwG regeneration will provide further insights into the regeneration of patient-specific SwGs and the development of potential intervention strategies for gland-derived diseases.
... growth factors, chemokines, hormones and androgens) (16). Niche and its components tightly regulate the behavior and function of stem cells through direct interactions and/or signaling cues from soluble factors (17). Traditionally, stem cell differentiation has been regulated through soluble signals such as growth factors. ...
... Therefore, rapid degradation should be avoided. To improve their mechanical integrity, natural biomaterials are often combined with synthetic ones to produce hybrid or composite biomaterials that achieve the advantages of both categories (17). ...
... However, synthetic materials often lack cell adhesion sites and cell recognition signals (24). Synthetic biomaterials can be obtained from Food and Drug Administration (FDA)-approved polymers with excellent biodegradable and biocompatible properties, such as poly (lactic acid) (PLA), polylactide caprolactone (PCL), polyglycolide (PGA) (17). Adapting synthetic materials is accomplished by adding biochemical modifications, modulating the material's mechanical properties, and/or determining the microscale structure and topography. ...
Article
Stem cells are cells that are not yet differentiated, can divide asymmetrically, differentiate into different cell types, and perform functional tissue repair. They are recognized as major cellular candidates for the regeneration of damaged tissues. Biomaterials and biomaterial scaffolds are essential in tissue engineering applications using stem cells. Recently, studies examining stem cell biomaterial interactions in different aspects have attracted attention. This review presents current information about the general properties of stem cells and biomaterials, stem cell-biomaterial interactions, three-dimensional (3D) tissue scaffolds used in stem cell studies, and 3D bioprinting.
... Those stem cells are routinely isolated, cultured, and amplified in vitro and relatedly applied. However, exogenous stem cell transplantation still has various disadvantages, for example, the complicated technology of in vitro operation, high-cost ex vivo cell culture, and even the potential ethical and safety risks Pacelli et al., 2017;Safina and Embree, 2022). ...
... Stem-cell-homing involves a series of physiological processes including cell recognition, migration, proliferation, and differentiation, and ultimately achieves tissue regeneration, which plays a huge role under certain conditions and has achieved remarkable therapeutic effects (Liesveld et al., 2020). In this method, biomaterials were utilized for bioactive factors delivery as well as the host's inherent regenerative potential activating (Safina and Embree, 2022;Xin et al., 2022;Yao and Lv, 2022;Zhao et al., 2022). By mobilizing appropriate stem/progenitor cells to specific spaces for tissue repair through cell-material interactions at the defect site, the endogenous regeneration process can be mimicked (Andreas et al., 2014;Mao et al., 2022). ...
... By mobilizing appropriate stem/progenitor cells to specific spaces for tissue repair through cell-material interactions at the defect site, the endogenous regeneration process can be mimicked (Andreas et al., 2014;Mao et al., 2022). The possible advantages of this strategy in promoting periodontal regeneration are as follows: firstly, it provides a solution to some of the limitations of stem cell transplantation, and transforms periodontal regeneration treatment methods into a clinically valid way; secondly, it gives full play to the potential of host self-repair and regeneration, making periodontal tissue regeneration safer; moreover, compared with the introduction of exogenous stem cells, it is simpler and less expensive to treat periodontal diseases and other diseases (Abdulghani and Mitchell, 2019; Safina and Embree, 2022). To make better use of the stem cell homing technique to restore the periodontal defect, this paper reviews the research status of the stem cell homing technique. ...
Article
Full-text available
The destruction of periodontal tissue is a crucial problem faced by oral diseases, such as periodontitis and tooth avulsion. However, regenerating periodontal tissue is a huge clinical challenge because of the structural complexity and the poor self-healing capability of periodontal tissue. Tissue engineering has led to advances in periodontal regeneration, however, the source of exogenous seed cells is still a major obstacle. With the improvement of in situ tissue engineering and the exploration of stem cell niches, the homing of endogenous stem cells may bring promising treatment strategies in the future. In recent years, the applications of endogenous cell homing have been widely reported in clinical tissue repair, periodontal regeneration, and cell therapy prospects. Stimulating strategies have also been widely studied, such as the combination of cytokines and chemokines, and the implantation of tissue-engineered scaffolds. In the future, more research needs to be done to improve the efficiency of endogenous cell homing and expand the range of clinical applications.
... ISTR refers to the use of engineering biomaterials carrying biological active molecules (drugs, growth factors, genes, polypeptides, and cells, etc.) to direct endogenous progenitor or stem cells to repair damaged tissues [13,14]. Compared with ex vivo systems, ISTR is relatively convenient and eliminates the need for harvested cells, making it more suitable for clinical translation [15,16]. For example, NeuraGen and Neurotube for nerve repair, INFUSE bone grafts for dental or orthopedic applications [13]. ...
... Angiogenesis is preferred for regeneration of vascularized tissues or organs such as heart and muscle [20], but should be inhibited during regeneration of avascular tissues such as cartilage and cornea [13,21]. To date, scaffolds including nanoparticles, fibers, hydrogels, microneedles (MNs), sponges and 3D printed scaffolds prepared from polymers, ceramics, metals and composites materials have been extensively investigated for ISTR [13,16]. Among them, MNs for minimally invasive delivery of therapeutic agents have become an emerging technology to promote tissue repair and healing, providing a novel development opportunity and challenge for ISTR. ...
Article
Full-text available
Tissue injury is a common clinical problem, which may cause great burden on patients' life. It is important to develop functional scaffolds to promote tissue repair and regeneration. Due to their unique composition and structure, microneedles have attracted extensive attention in various tissues regeneration, including skin wound, corneal injury, myocardial infarction, endometrial injury, and spinal cord injury et al. Microneedles with micro-needle structure can effectively penetrate the barriers of necrotic tissue or biofilm, therefore improving the bioavailability of drugs. The use of microneedles to deliver bioactive molecules, mesenchymal stem cells, and growth factors in situ allows for targeted tissue and better spatial distribution. At the same time, microneedles can also provide mechanical support or directional traction for tissue, thus accelerating tissue repair. This review summarized the research progress of microneedles for in situ tissue regeneration over the past decade. At the same time, the shortcomings of existing researches, future research direction and clinical application prospect were also discussed.
... During the exploration of perfect wound healing, scientists proposed the concept of endogenous skin regeneration. The main idea of endogenous skin regeneration is to implant bioactive biomaterials to the wound sites and take advantage of in vivo microenvironment, guiding the endogenous cells to regenerate skin tissues in situ [2]. In this process, cell-free biomaterials act as tissue scaffolds or vehicles for the recruitment, attachment, migration and differentiation of host cells. ...
Article
Full-text available
Endogenous regeneration is becoming an increasingly important strategy for wound healing as it facilitates skin's own regenerative potential for self-healing, thereby avoiding the risks of immune rejection and exogenous infection. However, currently applied biomaterials for inducing endogenous skin regeneration are simplistic in their structure and function, lacking the ability to accurately mimic the intricate tissue structure and regulate the disordered microenvironment. Novel biomimetic biomaterials with precise structure, chemical composition, and biophysical properties offer a promising avenue for achieving perfect endogenous skin regeneration. Here, we outline the recent advances in biomimetic materials induced endogenous skin regeneration from the aspects of structural and functional mimicry, physiological process regulation, and biophysical property design. Furthermore, novel techniques including in situ reprograming, flexible electronic skin, artificial intelligence , single-cell sequencing, and spatial transcriptomics, which have potential to contribute to the development of biomimetic biomaterials are highlighted. Finally, the prospects and challenges of further research and application of biomimetic biomaterials are discussed. This review provides reference to address the clinical problems of rapid and high-quality skin regeneration.
... Several studies investigated appropriate biomaterials that can provide mechanical, biological, topographical, and morphological properties to stimulate tissue regeneration [1,23,24]. The efficacy of an enormous variety of natural, synthetic, and hybrid biomaterials has been demonstrated for tissue regeneration [23,[25][26][27][28][29][30][31][32]. Fig. 2 shows the advantages and disadvantages of different biomaterials for tissue engineering. ...
... Among all of them, biomaterials are the most widely used in the medical field due to several important properties such as biocompatibility, bioactivity, degradability, long-term stability and many other important properties that make biomaterials able to adapt to the environment and organs in which they are implanted as a medical treatment, they can also remain in place for a long time without reducing their activity [4,5]. And, another property that distinguishes them from other types of advanced materials is that they can be designed to have a desired activation time as cells of the organs or the body that they implanted regenerate, they also deteriorate and disappear so that they do not have to be removed again [6]. ...
Article
Full-text available
Biomaterials are listed in advanced materials that have high biocompatibility which can easily adapt to the system in which they are implanted without leaving any adverse reactions and side effects. Due to their interesting properties such as biocompatibility, bioactivity, degradability, long-term stability, and many other important properties, all four main types of biomaterials (Bioceramics, Metallic biomaterials, Biopolymers, and Biocomposites) can be used in the medical field, either for medical treatment by implanting them in the human body, or the manufacturing of advanced medical devices. In this review, a comprehensive introduction to biomaterials has been mentioned. Also, the general properties of biomaterials are explained especially these interesting properties that are helpful to use in the medical field. And finally, the medical applications of each of the different types of biomaterials have been reviewed.
... In situ tissue regeneration proposes a new conception of recruiting and utilizing the resident stem cells of the host for tissue regeneration (Safina and Embree, 2022). To this end, stromal cell-derived factor-1 (SDF-1) from the CXC chemokine family plays a key role in recruiting CXCR4 (CXC motif chemokine receptor type 4)-positive stem and progenitor cells in the early stage of tissue repairment (Yu et al., 2020;Ling et al., 2022). ...
Article
Full-text available
Purpose: Poly (lactic-co-glycolic acid)-based nanoparticles (PLGA NPs) have been widely used as the carrier for sustainable drug delivery. However, the drug release from the NPs was usually incomplete and uncontrollable. Herein, a low intensity pulsed ultrasound (LIPUS) assisted SDF-1/BMP-2@nanoparticles (S/B@NPs) system was fabricated to facilitate stem cell recruitment-osteogenesis for periodontal bone regeneration. Methods: In this work, S/B@NPs were prepared with double-emulsion synthesis method. Then the S/B release profile from NPs was evaluated with or without low intensity pulsed ultrasound treatment. Afterwards, the stem cell recruiting and osteoinductive capacities of LIPUS-S/B@NPs were detected with human periodontal ligament cells (hPDLCs) in vitro and in a rat periodontal bone defect model. Results: The results indicated that S/B@NPs were successfully prepared and LIPUS could effectively regulate the release of S/B and increase their final releasing amount. Moreover, LIPUS-S/B@NPs system significantly promoted hPDLCs migrating and osteogenesis in vitro and recruiting rBMSCs to the rat periodontal defect and facilitated bone regeneration in vivo. Conclusion: Our LIPUS assisted S/B@NPs system can effectively facilitate stem cell recruitment and periodontal bone regeneration. Considering its reliable safety and therapeutic effect on bone fracture, LIPUS, as an adjuvant therapy, holds great potential in the regulation of drug delivery systems for bone healing.
... 2 The outstanding advantages of hydrogels 2.1 Physicochemical properties similar to the extracellular matrix of natural tissue Tissue repair and regeneration cannot be achieved without cell growth, reproduction and differentiation. Cells can sense changes in the surrounding extracellular matrix and perform the corresponding biological responses (Safina and Embree, 2022). The structure, stiffness, degradability, pore size and other physicochemical properties of hydrogels are similar to those of the extracellular matrix of natural tissues, which can provide a good microenvironment for cell proliferation and differentiation. ...
Article
Full-text available
Tissue defects can be accompanied by functional impairments that affect the health and quality of life of patients. Hydrogels are three-dimensional (3D) hydrophilic polymer networks that can be used as bionic functional tissues to fill or repair damaged tissue as a promising therapeutic strategy in the field of tissue engineering and regenerative medicine. This paper summarises and discusses four outstanding advantages of hydrogels and their applications and advances in the repair and regeneration of tissue defects. First, hydrogels have physicochemical properties similar to the extracellular matrix of natural tissues, providing a good microenvironment for cell proliferation, migration and differentiation. Second, hydrogels have excellent shape adaptation and tissue adhesion properties, allowing them to be applied to a wide range of irregularly shaped tissue defects and to adhere well to the defect for sustained and efficient repair function. Third, the hydrogel is an intelligent delivery system capable of releasing therapeutic agents on demand. Hydrogels are capable of delivering therapeutic reagents and releasing therapeutic substances with temporal and spatial precision depending on the site and state of the defect. Fourth, hydrogels are self-healing and can maintain their integrity when damaged. We then describe the application and research progress of functional hydrogels in the repair and regeneration of defects in bone, cartilage, skin, muscle and nerve tissues. Finally, we discuss the challenges faced by hydrogels in the field of tissue regeneration and provide an outlook on their future trends.
... However, the explanation may also be attributed to the rearrangement of cytoskeletal networks after cell-receptor recognition and aggregation. For instance, integrin, once bound to the surface of biomaterials, may, in turn, activate the downstream Wnt, YAP and c-Jun N-terminal kinase signaling, leading to changes in gene expression (57). The design of iBTE scaffolds aims to create a bioinstructive microenvironment to regulate the behavior of endogenous cells through materials, which requires a comprehensive understanding of organismal physiopathology, cellular function and material science. ...
Article
Full-text available
Numerous physiological processes occur following bone fracture, including inflammatory cell recruitment, vascularization, and callus formation and remodeling. In particular circumstances, such as critical bone defects or osteonecrosis, the regenerative microenvironment is compromised, rendering endogenous stem/progenitor cells incapable of fully manifesting their reparative potential. Consequently, external interventions, such as grafting or augmentation, are frequently necessary. In situ bone tissue engineering (iBTE) employs cell-free scaffolds that possess microenvironmental cues, which, upon implantation, redirect the behavior of endogenous stem/progenitor cells towards a pro-regenerative inflammatory response and reestablish angiogenesis-osteogenesis coupling. This process ultimately results in vascularized bone regeneration (VBR). In this context, a comprehensive review of the current techniques and modalities in VBR-targeted iBTE technology is provided.
... In situ cartilage regeneration by cell-free scaffolds via recruiting endogenous stem cells has been thrown into renewed focus. 18 Multiple mesenchymal stem/progenitor cells (MSPCs) are present in joint-resident niches, such as bone marrow-derived MSCs (BMSCs), synovium-derived MSCs, synovium fluid-derived MSCs, and cartilage-derived progenitor cells, which are capable of migrating to damaged sites and participating in cartilage regeneration. 19 Damaged cartilage can release many injuryassociated products as natural recruitment signals to recruit surrounding MSPCs, nonetheless, these signals are too weak to recruit adequate endogenous cells. ...
Article
Articular cartilage is refractory to self-healing due to the absence of vascular, nervous, and lymphatic systems, and its repair remains a clinical challenge. Tissue regeneration through in situ recruitment of stem cells via cell-free scaffolds is a promising alternative strategy. Herein, a kind of functional injectable hydrogel system (Col-Apt@KGN MPs), which is a collagen-based and microsphere-embedded cell-free scaffold, was designed to achieve spatiotemporal regulation of endogenous mesenchymal stem cells (MSCs) recruitment and their chondrogenic differentiation by respective release of aptamer 19S (Apt19S) and kartogenin (KGN). In vitro results confirmed that the Col-Apt@KGN MPs hydrogel had sequential release characteristics. Apt19S was rapidly released from the hydrogel within 6 days, while KGN was slowly released for 33 days via the degradation of poly(lactic-co-glycolic acid) (PLGA) microspheres. When cultured with MSCs, the Col-Apt@KGN MPs hydrogel supported the adhesion, proliferation, and chondrogenic differentiation of MSCs. In vivo results indicated that the Col-Apt@KGN MPs hydrogel effectively promoted the recruitment of endogenous MSCs in a rabbit full-thickness cartilage defect model; furthermore, the Col-Apt@KGN MPs hydrogel enhanced the secretion of cartilage specific extracellular matrix and achieved the reconstruction of subchondral bone. This study demonstrates that the Col-Apt@KGN MPs hydrogel possesses great potential in recruitment of endogenous stem cells and cartilage tissue regeneration.
... Adult stem cells possess self-renewal and pluripotent properties, making them ideal candidates for cell therapeutic applications aimed at facilitating tissue regeneration and repair. Adipose-derived stem cells (ADSCs) are particularly useful in research [1,2] due to their abundance and ease of acquisition through minimally invasive procedures [3]. ADSCs are highly pluripotent and can differentiate into multiple lineages, including adipogenic, osteogenic, chondrogenic, neural, hepatic, and myogenic lineages, making them a compelling choice for therapeutic applications [4,5]. ...
Article
Full-text available
Hydrogels are widely used in stem cell therapy due to their extensive tunability and resemblance to the extracellular matrix (ECM), which has a three-dimensional (3D) structure. These features enable various applications that enhance stem cell maintenance and function. However, fast and simple hydrogel fabrication methods are desirable for stem cells for efficient encapsulation and to reduce adverse effects on the cells. In this study, we present a one-pot double-crosslinked hydrogel consisting of polyethylene glycol (PEG) and collagen, which can be prepared without the multi-step sequential synthesis of each network, by using bio-orthogonal chemistry. To enhance the adipogenic differentiation efficiency of adipose-derived stem cells (ADSCs), we added degradable components within the hydrogel to regulate matrix stiffness through cell-mediated degradation. Bio-orthogonal reactions used for hydrogel gelation allow rapid gel formation for efficient cell encapsulation without toxic by-products. Furthermore, the hybrid network of synthetic (PEG) and natural (collagen) components demonstrated adequate mechanical strength and higher cell adhesiveness. Therefore, ADSCs grown within this hybrid hydrogel proliferated and functioned better than those grown in the single-crosslinked hydrogel. The degradable elements further improved adipogenesis in ADSCs with dynamic changes in modulus during culture and enabled the retrieval of differentiated cells for potential future applications.
... Biomaterials hold exceptional promise for the manufacturing and delivery of cellular therapeutics due to their biocompatibility and tunable mechanical properties, degradation rate, charge, and microstructure. [1][2][3][4][5][6] Current applications of biomaterial hydrogels and scaffolds include tissue engineered constructs, 3,7,8 drug delivery depots, 9,10 and vehicles for cellular proliferation, differentiation, and delivery. 3,11,12 Biomaterial scaffolds made from sodium alginate hold promise as delivery vehicles and 3-D structures to organize cells. ...
Article
Developing the next generation of cellular therapies will depend on fast, versatile, and efficient cellular reprogramming. Novel biomaterials will play a central role in this process by providing scaffolding and...
... Biomaterials hold exceptional promise for the manufacturing and delivery of cellular therapeutics due to their biocompatibility and tunable mechanical properties, degradation rate, charge, and microstructure. [1][2][3][4][5][6] Current applications of biomaterial hydrogels and scaffolds include tissue engineered constructs, 3,7,8 drug delivery depots, 9,10 and vehicles for cellular proliferation, differentiation, and delivery. 3,11,12 Biomaterial scaffolds made from sodium alginate hold promise as delivery vehicles and 3-D structures to organize cells. ...
... Cell-free therapy based on a combination of biomaterials and endogenous stem cells is a promising approach to treatment in the field of tissue regeneration (Ng et al., 2021). Studies have shown that endogenous stem cells, such as BMSCs, play an important role in cartilage regeneration (Safina & Embree, 2022). One reason regenerative hyaline cartilage turns into fibrocartilage is that the damaged area cannot recruit sufficient stem cells. ...
Article
Regeneration of injured articular cartilage is limited by low early-stage recruitment of stem cells and insufficient chondrogenic differentiation. Hydrogels are widely used to repair cartilage because they have excellent mechanical and biological properties. In this study, a dual drug-loaded thermosensitive hydroxypropyl chitin hydrogel (HPCH) system was prepared to release stromal-derived factor-1α-like polypeptides (SDFP) and kartogenin (KGN) for stem-cell recruitment and chondrogenic differentiation. The hydrogel had a network structure that promoted cell growth and nutrient exchange. Moreover, it was temperature sensitive and suitable for filling irregular defects. The system showed good biocompatibility in vitro and promoted stem-cell recruitment and chondrogenic differentiation. Furthermore, it reduced chondrocyte catabolism under inflammatory conditions. Animal experiments demonstrated that the dual-drug hydrogel systems can promote the regeneration of articular cartilage in rats. This study confirmed that an HPCH system loaded with KGN and SDFP could effectively repair articular cartilage defects and represents a viable treatment strategy.
... The produced tissue is then implanted into the desired tissue, so it must be of the same size and shape as the defect area, the scaffold degrades over time to permit the substitution with the newly regenerated tissues [20][21][22][23]. This approach presents scaffolds of good mechanical properties and allows the use of many biomaterials [24][25][26][27][28]; however, it requires sophisticated optimization of the conditions in the bioreactors to allow initial cell proliferation, high cost, donor site morbidity, and rejection of the implanted tissue may occur [29]. While (ii) in situ tissue engineering represents a simple and convenient solution that involves the pre-fabrication of a scaffold made from biocompatible biomaterials with a specific size and shape and its implantation directly into the required tissue without the need for prior seeding with cells, it relies on attracting the surrounding cells by promoting the host's tissue regeneration [8,14,15,30,31]. ...
Article
Full-text available
Tissue engineering has emerged as an interesting field nowadays; it focuses on accelerating the auto-healing mechanism of tissues rather than organ transplantation. It involves implanting an In Vitro cultured initiative tissue or a scaffold loaded with tissue regenerating ingredients at the damaged area. Both techniques are based on the use of biodegradable , biocompatible polymers as scaffolding materials which are either derived from natural (e.g. alginates, celluloses, and zein) or synthetic sources (e.g. PLGA, PCL, and PLA). This review discusses in detail the recent applications of different biomaterials in tissue engineering highlighting the targeted tissues besides the in vitro and in vivo key findings. As well, smart biomaterials (e.g. chitosan) are fascinating candidates in the field as they are capable of elucidating a chemical or physical transformation as response to external stimuli (e.g. temperature, pH, magnetic or electric fields). Recent trends in tissue engineering are summarized in this review highlighting the use of stem cells, 3D printing techniques, and the most recent 4D printing approach which relies on the use of smart biomaterials to produce a dynamic scaffold resembling the natural tissue. Furthermore, the application of advanced tissue engineering techniques provides hope for the researchers to recognize COVID-19/host interaction, also, it presents a promising solution to rejuvenate the destroyed lung tissues. Graphical abstract
Article
Stem cell-based therapy is a novel technique used to reconstruct the ocular surface of patients with limbal stem cell deficiency (LSCD). However, the reported stem cell therapies for LSCD are limited to two-dimensional (2D) culture systems, which cannot mimic the native microenvironment in which stem cells reside in vivo. Herein, an injectable photocrosslinkable hydrogel was developed based on porous gelatin methacryloyl (GelMA)/silk fibroin glycidyl methacrylate (SilMA), which encapsulated limbal stem cells (LSCs) to reconstruct ocular surfaces. The porous GelMA/SilMA hydrogels demonstrated excellent transparency and good adhesion. More importantly, this hydrogel maintained viability, supported the adhesion of encapsulated rabbit LSCs and promoted their migration in vitro in three-dimensional (3D) culture environments. Proteomic analysis confirmed that the porous GelMA/SilMA hydrogel 3D cell culture system maintained both the self-renewal and differentiation of the LSCs. Furthermore, histological analyses demonstrated that the porous GelMA/SilMA hydrogel encapsulating LSCs effectively promoted the re-epithelization of LSCD. Porous hydrogels were subjected to simplified serial cell culture operations to construct an ocular surface with a uniform cell distribution. These findings provide valuable insights into stem cell therapies and regenerative medicine for ocular surface reconstruction.
Article
Full-text available
Cardiovascular diseases continue to challenge global health, demanding innovative therapeutic solutions. This review delves into the transformative role of mesenchymal stem cells (MSCs) in advancing cardiovascular therapeutics. Beginning with a historical perspective, we trace the development of stem cell research related to cardiovascular diseases, highlighting foundational therapeutic approaches and the evolution of cell-based treatments. Recognizing the inherent challenges of MSC-based cardiovascular therapeutics, which range from understanding the pro-reparative activity of MSCs to tailoring patient-specific treatments, we emphasize the need to refine the pro-regenerative capacity of these cells. Crucially, our focus then shifts to the strategies of the fourth generation of cell-based therapies: leveraging the secretomic prowess of MSCs, particularly the role of extracellular vesicles; integrating biocompatible scaffolds and artificial sheets to amplify MSCs’ potential; adopting three-dimensional ex vivo propagation tailored to specific tissue niches; harnessing the promise of genetic modifications for targeted tissue repair; and institutionalizing good manufacturing practice protocols to ensure therapeutic safety and efficacy. We conclude with reflections on these advancements, envisaging a future landscape redefined by MSCs in cardiovascular regeneration. This review offers both a consolidation of our current understanding and a view toward imminent therapeutic horizons.
Article
Macrophages play a prominent role in tissue development, homeostasis, and repair. In addition, their fundamental and well-known functions are integral to the host’s defense system, including immune responses and debris removal from apoptotic cells. These immune cells are remarkably responsive to various stimuli in the microenvironment, inducing multiple forms of differentiation with distinct phenotypes and roles. These stimuli comprise biochemical signals and physical biomaterial properties. Thus, understanding how immune cells differentiate is crucial for designing biomaterials, tissue scaffolds, implantable materials, and regenerative medicine, such as developing new drugs and immunomodulatory drug delivery systems. This review discusses about the latest techniques in inducing immune cell differentiation.
Article
Full-text available
The prevalence of infertility caused by endometrial defects is steadily increasing, posing a significant challenge to women's reproductive health. In this study, injectable “homing‐like” bioactive decellularized extracellular matrix short‐fibers (DEFs) of porcine skin origin are innovatively designed for endometrial and fertility restoration. The DEFs can effectively bind to endometrial cells through noncovalent dipole interactions and release bioactive growth factors in situ. In vitro, the DEFs effectively attracted endometrial cells through the “homing‐like” effect, enabling cell adhesion, spreading, and proliferation on their surface. Furthermore, the DEFs effectively facilitated the proliferation and angiogenesis of human primary endometrial stromal cells (HESCs) and human umbilical vein endothelial cells (HUVECs), and inhibited fibrosis of pretreated HESCs. In vivo, the DEFs significantly accelerated endometrial restoration, angiogenesis, and receptivity. Notably, the deposition of endometrial collagen decreased from 41.19 ± 2.16% to 14.15 ± 1.70% with DEFs treatment. Most importantly, in endometrium‐injured rats, the use of DEFs increased the live birth rate from 30% to an impressive 90%, and the number and development of live births close to normal rats. The injectable “homing‐like” bioactive DEFs system can achieve efficient live births and intrauterine injection of DEFs provides a new promising clinical strategy for endometrial factor infertility.
Article
Full-text available
Biotechnology constitutes a realm of inquiry that extends its benefits across diverse domains of knowledge, ranging from agricultural sciences to clinical applications. This field leverages technology to address challenges frequently entailing living organisms. Of current significance is the endeavor to elucidate the process of animal regeneration, particularly within the human species, given its burgeoning potential as an ally in the treatment and cure of various maladies. Recognizing the societal importance of this subject, the present article seeks to expound upon contemporary biotechnological advancements facilitating the exploration of cellular and tissue regeneration for the treatment of human diseases. To this end, a comprehensive review of articles delineating the current landscape was conducted, involving a comparative analysis of regenerative activity across species, with a specific focus on humans. Evidentially, the use of biomaterials in tissue regeneration assumes paramount importance, albeit not without the formidable challenge posed by the inflammatory process. Stem cells, conversely, present themselves as promising entities in the realm of regeneration. However, their interaction within the host organism necessitates further scrutiny to attain a more nuanced understanding. Despite strides made in the field of regenerative medicine, the lack of comprehensive comprehension regarding the properties of biomaterials and their responses within the human body constrains their clinical applicability. Nevertheless, an auspicious future is envisioned, marked by advancements in biomaterials and a heightened understanding of interactions within the human body, thereby fostering the development of more efficacious treatments for a myriad of diseases.
Article
Full-text available
Injectable biomaterials have garnered increasing attention for their potential and beneficial applications in minimally invasive surgical procedures and tissue regeneration. Extracellular matrix (ECM) hydrogels and porous synthetic polymer microspheres can be prepared for injectable administration to achieve in situ tissue regeneration. However, the rapid degradation of ECM hydrogels and the poor injectability and biological inertness of most polymeric microspheres limit their pro-regenerative capabilities. Here, we develop a biomaterial system consisting of elastic porous poly(l-lactide-co-ε-caprolactone) (PLCL) microspheres mixed with ECM hydrogels as injectable composites with interleukin-4 (IL-4) and insulin-like growth factor-1 (IGF-1) dual-release functionality. The developed multifunctional composites have favorable injectability and biocompatibility, and regulate the behavior of macrophages and myogenic cells following injection into muscle tissue. The elicited promotive effects on tissue regeneration are evidenced by enhanced neomusle formation, vascularization, and neuralization at 2-months post-implantation in a male rat model of volumetric muscle loss. Our developed system provides a promising strategy for engineering bioactive injectable composites that demonstrates desirable properties for clinical use and holds translational potential for application as a minimally invasive and pro-regenerative implant material in multiple types of surgical procedures.
Article
Full-text available
Abstract Endogenous stem cell (ESC) exploration refers to an increase in the number of stem cells residing in the human body. This can be achieved by administering natural substances, such as honey, which can activate stem cells. Numerous studies have explored the potential of honey for animal and human health, including its antibacterial, antifungal, antiparasitic, anti-inflammatory, immunomodulatory, antioxidant, anti-osteoporosis, anti-malnutrition, and anti-fertility properties. Here, we review the scientific data on the importance of honey as a potential natural substance, and how its ability to activate endogenous stem cells can lead to new therapeutic strategies for different diseases. Honey majorly contributes to stem cell proliferation and differentiation and consequently tissue repair via mobilization and homing of endogenous stem cells toward damaged tissues. Altogether, bioactive compounds in honey or phytochemicals are potential compounds for enhancing stem cell therapeutics for several degenerative diseases.
Article
Full-text available
Stem cell-based regenerative therapies, which harness the self-renewal and differentiation property of stem cells, have been in the spotlight due to their widespread applications in treating degenerative, aging and other, generally intractable diseases. Therapeutically effective hematopoietic stem cells, mesenchymal stem cells, embryonic stem cells and induced pluripotent stem cells have been used in numerous basic and translational studies with exciting results. However, pre-/post-transplantation issues of poor cell survival and retention, uncontrolled differentiation and insufficient numbers of cells engrafted into host tissues are the major challenges in stem cell-based regenerative therapies. Engineered biomaterials have adjustable biochemical and biophysical properties that significantly affect cell behaviours, such as cell engraftment, survival, migration and differentiation outcomes, thereby enhancing the engraftment of implanted stem cells and guide tissue regeneration. Therefore, the combination of stem cell biology with bioengineered materials is a promising strategy to improve the therapeutic outcomes of stem cell-based regenerative therapy. In this review, we summarize the advances in the modulation of behaviours of stem cells via engineered biomaterials. We then present different approaches in harnessing bioengineered materials to enhance transplantation of stem cells. Finally, we will provide future directions in regenerative therapy using stem cells.
Article
The repair of large-size cranial bone defects caused by traumatic brain injury (TBI) remains a substantial clinical challenge. On one hand, traditional bone implants with intrinsic brittle and poor recovery features hinder their immediate implantation for cranioplasty applications. On the other hand, using exogenous growth factors to enhance the osteo-bioactivity of bone implants often leads to efficacy, safety, and cost concerns. Thus far, the authors develop a growth factor-free pliable hydrogel with multiple functions for mediating endogenous growth factor production and stem cell functions in cranioplasty. The pliable hydrogels are based on GelMA networks, in which the mechanical properties and protein affinity were strengthened by the crosslinked poly (ethylene glycol) disuccinimidyl succinate (PEG-(SS)2), while the antioxidant capability and osteoinductivity were remarkedly enhanced through the decoration of magnesium-seamed C-propylpyrogallol[4]arene cages (PgC3Mg). In vitro and in vivo results confirmed that the versatile hydrogel with excellent biocompatibility and biodegradability can improve osteogenic differentiation and cranial bone regeneration by facilitating growth factor production, endogenous cell recruitment and angiogenesis. These findings indicate that the versatile hydrogels represent a potential avenue for developing growth factor-free pliable scaffolds in cranioplasty after TBI.
Article
Full-text available
The aberrant mechanical microenvironment in degenerated tissues induces misdirection of cell fate, making it challenging to achieve efficient endogenous regeneration. Herein, a hydrogel microsphere-based synthetic niche with integrated cell recruitment and targeted cell differentiation properties via mechanotransduction was constructed for enhanced endogenous regeneration. Through the incorporation of microfluidics and photo-polymerization strategies, we prepared fibronectin (Fn) modified methacrylated gelatin (GelMA) microspheres with the independently tunable elastic modulus(1-10Kpa) and ECM ligand density (2 and 10 μg/ml), allowing a wide range of cytoskeleton modulation to trigger the corresponding mechanobiological signaling to direct cell fate. The combination of the soft matrix (2Kpa) and low ligand density (2 μg/ml) could support the nucleus pulposus (NP)-like differentiation of intervertebral disc (IVD) progenitor/stem cells by translocating Yes-associated protein (YAP), without the addition of inducible biochemical factors. Meanwhile, platelet-derived growth factor-BB was loaded onto Fn-GelMA microspheres (PDGF@Fn-GelMA) via the heparin-binding domain of Fn and exhibited continuous release for over 28 days to initiate endogenous cell recruitment. In in vivo experiments, hydrogel microsphere-niche maintained the IVD structure and stimulated ECM synthesis, thereby enhancing the endogenous regeneration of NP. Overall, this synthetic niche with cell recruiting and mechanical training capabilities offered a promising novel strategy for endogenous tissue regeneration. This article is protected by copyright. All rights reserved.
Preprint
Full-text available
Injectable biomaterials have garnered increasing attention for their potential and beneficial applications in minimally invasive surgical procedures and tissue regeneration. Extracellular matrix (ECM) hydrogels and porous synthetic polymer microspheres can be prepared for injectable administration to achieve in situ tissue regeneration. However, the rapid degradation of ECM hydrogels and the poor injectability and biological inertness of most polymeric microspheres limit their pro-regenerative capabilities. Here, we developed a biomaterial system consisting of elastic porous poly(l-lactide-co-ε-caprolactone) (PLCL) microspheres mixed with ECM hydrogels as injectable composites with interleukin-4 (IL-4) and insulin-like growth factor-1 (IGF-1) dual-release functionality. The developed multifunctional composites had favorable injectability and biocompatibility, and regulated the behavior of macrophages and myogenic cells following injection into muscle tissue. The elicited promotive effects on tissue regeneration were evidenced by enhanced neomusle formation, vascularization, and neuralization at 2-months post-implantation in a rat model of volumetric muscle loss. Our developed system provides a promising strategy for engineering bioactive injectable composites that demonstrates desirable properties for clinical use and holds translational potential for application as a minimally invasive and pro-regenerative implant material in multiple types of surgical procedures.
Article
The excessive reactive oxygen species (ROS) level, inflammation, and weak tissue regeneration ability after annulus fibrosus (AF) injury constitute an unfavorable microenvironment for AF repair. AF integrity is crucial for preventing disc herniation after discectomy; however, there is no effective way to repair the AF. Herein, a composite hydrogel integrating properties of antioxidant, anti-inflammation, and recruitment of AF cells is developed through adding mesoporous silica nanoparticles modified by ceria and transforming growth factor β3 (TGF-β3) to the hydrogels. The nanoparticle loaded gelatin methacrylate/hyaluronic acid methacrylate composite hydrogels eliminate ROS and induce anti-inflammatory M2 type macrophage polarization. The released TGF-β3 not only plays a role in recruiting AF cells but is also responsible for promoting extracellular matrix secretion. The composite hydrogels can be solidified in situ in the defect area to effectively repair AF in rats. The strategies targeting endogenous ROS removal and improving the regenerative microenvironment by the nanoparticle-loaded composite hydrogels have potential applications in AF repair and intervertebral disc herniation prevention.
Article
Compared with most nondegradable or slowly degradable bone repair materials, bioactive biodegradable porous scaffolds with certain mechanical strengths can promote the regeneration of both new bone and vasculature while the cavity created by their degradation can be replaced by the infiltration of new bone tissue. Mineralized collagen (MC) is the basic structural unit of bone tissue, and silk fibroin (SF) is a natural polymer with adjustable degradation rates and superior mechanical properties. In this study, a three-dimensional porous biomimetic composite scaffold with a two-component SF-MC system was constructed based on the advantages of both materials. The spherical mineral agglomerates of the MC were uniformly distributed on the surface and inside the SF skeleton, which ensured good mechanical properties while regulating the degradation rate of the scaffold. Second, the SF-MC scaffold had good osteogenic induction of bone marrow mesenchymal stem cells (BMSCs) and preosteoblasts (MC3T3-E1) and also promoted the proliferation of MC3T3-E1 cells. Finally, in vivo 5 mm cranial defect repair experiments confirmed that the SF-MC scaffold stimulated vascular regeneration and promoted new bone regeneration in vivo by means of in situ regeneration. Overall, we believe that this low-cost biomimetic biodegradable SF-MC scaffold with many advantages has some clinical translation prospects.
Article
Bone defects are the second most common tissue grafts after blood. However, bone grafts face several problems, such as bone scaffolds, which have low bioactivity and are prone to corrosion. Much of the current research on bone scaffolds is focused on the mechanical aspects such as structure and strength. Surface modification of the bone scaffold is carried out in terms of the mechanical structure or structural design of the bone scaffold with reference to a bionic structure. However, with the development of mechanical designs, materials science, and medicine, many studies have reported that promoting bone growth by modifying the structure of the scaffold or coating is not possible. Therefore, the application of a bioactive coating to the surface of the bone scaffold is particularly important to generate a synergistic effect between the structure and active coating. In this article, we present several perspectives to improve the bioactivity of bone scaffolds, including corrosion resistance, loading of bioactive coatings or drugs on bone scaffolds, improved adhesion to the surface of the bone scaffolds, immune response modulation, and drawing on bionic structures during manufacturing.
Article
Full-text available
Background Because some of its CNS neurons (e.g., retinal ganglion cells after optic nerve crush (ONC)) regenerate axons throughout life, whereas others (e.g., hindbrain neurons after spinal cord injury (SCI)) lose this capacity as tadpoles metamorphose into frogs, the South African claw-toed frog, Xenopus laevis, offers unique opportunities for exploring differences between regenerative and non-regenerative responses to CNS injury within the same organism. An earlier, three-way RNA-seq study (frog ONC eye, tadpole SCI hindbrain, frog SCI hindbrain) identified genes that regulate chromatin accessibility among those that were differentially expressed in regenerative vs non-regenerative CNS [11]. The current study used whole genome bisulfite sequencing (WGBS) of DNA collected from these same animals at the peak period of axon regeneration to study the extent to which DNA methylation could potentially underlie differences in chromatin accessibility between regenerative and non-regenerative CNS. Results Consistent with the hypothesis that DNA of regenerative CNS is more accessible than that of non-regenerative CNS, DNA from both the regenerative tadpole hindbrain and frog eye was less methylated than that of the non-regenerative frog hindbrain. Also, consistent with observations of CNS injury in mammals, DNA methylation in non-regenerative frog hindbrain decreased after SCI. However, contrary to expectations that the level of DNA methylation would decrease even further with axotomy in regenerative CNS, DNA methylation in these regions instead increased with injury. Injury-induced differences in CpG methylation in regenerative CNS became especially enriched in gene promoter regions, whereas non-CpG methylation differences were more evenly distributed across promoter regions, intergenic, and intragenic regions. In non-regenerative CNS, tissue-related (i.e., regenerative vs. non-regenerative CNS) and injury-induced decreases in promoter region CpG methylation were significantly correlated with increased RNA expression, but the injury-induced, increased CpG methylation seen in regenerative CNS across promoter regions was not, suggesting it was associated with increased rather than decreased chromatin accessibility. This hypothesis received support from observations that in regenerative CNS, many genes exhibiting increased, injury-induced, promoter-associated CpG-methylation also exhibited increased RNA expression and association with histone markers for active promoters and enhancers. DNA immunoprecipitation for 5hmC in optic nerve regeneration found that the promoter-associated increases seen in CpG methylation were distinct from those exhibiting changes in 5hmC. Conclusions Although seemingly paradoxical, the increased injury-associated DNA methylation seen in regenerative CNS has many parallels in stem cells and cancer. Thus, these axotomy-induced changes in DNA methylation in regenerative CNS provide evidence for a novel epigenetic state favoring successful over unsuccessful CNS axon regeneration. The datasets described in this study should help lay the foundations for future studies of the molecular and cellular mechanisms involved. The insights gained should, in turn, help point the way to novel therapeutic approaches for treating CNS injury in mammals.
Article
Full-text available
Since Paul Ehrlich’s introduction of the “magic bullet” concept in 1908¹, drug developers have been seeking new ways to target drug activity to diseased cells while limiting effects on normal tissues. In recent years, it has been proposed that coupling riboswitches capable of detecting RNA biomarkers2, 3 to small interfering RNAs (siRNAs) to create siRNA pro-drugs could selectively activate RNA interference (RNAi) activity in specific cells⁴⁻⁷. However, this concept has not previously been achieved. We report here that we have accomplished this goal, validating a simple and programmable new design that functions reliably in mammalian cells. We show that these conditionally activated siRNAs (Cond-siRNAs) can switch RNAi activity against different targets between clearly distinguished OFF and ON states in response to different cellular RNA biomarkers. Notably, in a rat cardiomyocyte cell line (H9C2)⁸, one version of our construct demonstrated biologically meaningful inhibition of a heart disease related target gene (PPP3CA)⁹ in response to increased expression of the pathological marker atrial natriuretic peptide (NPPA) messenger RNA (mRNA). Our results demonstrate the ability of synthetic riboswitches to regulate gene expression in mammalian cells, opening a new path for development of programmable siRNA pro-drugs.
Article
Full-text available
Background: A tendency towards extensive regional lymph node metastasis (LNM) is a typical clinical characteristic of esophageal squamous cell carcinoma (ESCC). Up-regulated microRNA (miR)-19a-3p was verified as a predictor of LNM in ESCC in previous microarray analyses, but the underlying mechanisms remain unclear. Here, in vitro experiments were performed to confirm the effect of miR-19a-3p on promoting LNM and to explore the underlying mechanisms. Methods: KYSE-150 and TE-1 cell lines were transfected with lentiviral vectors to inhibit miR-19a-3p (LV-miR-19a-3p-inhibition), and cell proliferation, invasion, and migration were assessed. Target genes of miR-19a-3p were identified by sequencing analysis and quantitative reverse transcription PCR (qRT-PCR); Western blotting was performed to confirm targets and explore the potential mechanisms underlying the effect of miR-19a-3p on LNM. Results: miR-19a-3p had no effect on ESCC cell proliferation, whereas miR-19a-3p overexpression promoted the invasion and migration of ESCC cells. qRT-PCR verification and western blot analysis showed that LV-miR-19a-3p-inhibition downregulated cell division cycle 42 (CDC42), Rac family small GTPase 1 (RAC1), and p21 activated kinase 1 (PAK1). Conclusions: Overexpression of miR-19a-3p increased the invasion and migration of ESCC cells via the RAC1/CDC42-PAK1 pathway, suggesting that this pathway mediates the effect of miR-19a-3p on promoting LNM in ESCC.
Article
Full-text available
Current treatment approaches toward spinal cord injuries (SCI) have mainly focused on overcoming the inhibitory microenvironment that surrounds lesion sites. Unfortunately, the mere modulation of the cell/tissue microenvironment is often insufficient to achieve desired functional recovery. Therefore, stimulating the intrinsic growth ability of injured neurons becomes crucial. MicroRNAs (miRs) play significant roles during axon regeneration by regulating local protein synthesis at growth cones. However, one challenge of using miRs to treat SCI is the lack of efficient delivery approaches. Here, a 3D fiber-hydrogel scaffold is introduced which can be directly implanted into a spinal cord transected rat. This 3D scaffold consists of aligned electrospun fibers which provide topographical cues to direct axon regeneration, and collagen matrix which enables a sustained delivery of miRs. Correspondingly, treatment with Axon miRs (i.e., a cocktail of miR-132/miR-222/miR-431) significantly enhances axon regeneration. Moreover, administration of Axon miRs along with anti-inflammatory drug, methylprednisolone, synergistically enhances functional recovery. Additionally, this combined treatment also decreases the expression of pro-inflammatory genes and enhance gene expressions related to extracellular matrix deposition. Finally, increased Axon miRs dosage with methylprednisolone, significantly promotes functional recovery and remyelination. Altogether, scaffold-mediated Axon miR treatment with methylprednisolone is a promising therapeutic approach for SCI.
Article
Full-text available
The extracellular microenvironment proved to exert a potent regulatory effect over different aspects of Embryonic Stem Cells (ESCs) behavior. In particular, the employment of engineered culture surfaces aimed at modulating ESC self-organization resulted effective in directing ESCs toward specific fate decision. ESCs fluctuate among different levels of functional potency and in this context the Zscan4 gene marks the so-called “metastate,” a cellular state in which ESCs retain both self-renewal and pluripotency capabilities. Here we investigated the impact of topographic cues on ESCs pluripotency, differentiation and organization capabilities. To this aim, we engineered culturing platforms of nanograted surfaces with different features size and we investigated their impact on ESCs multicellular organization and Zscan4 gene expression. We showed that the morphology of ESC-derived aggregates and Zscan4 expression are strictly intertwined. Our data suggest that ESC Zscan4 metastate can be promoted if the adhesive surface conditions guide cellular self-aggregation into 3D dome-like structure, in which both cell-material interactions and cell-cell contact are supportive for Zscan4 expression.
Article
Full-text available
Background: Tissue engineering of the annulus fibrosus (AF) shows promise as a treatment for patients with degenerative disc disease (DDD). However, it remains challenging due to the intrinsic heterogeneity of AF tissue. Fabrication of scaffolds recapitulating the specific cellular, componential, and microstructural features of AF, therefore, is critical to successful AF tissue regeneration. Methods: Poly-L-lactic acid (PLLA) fibrous scaffolds with various fiber diameters and orientation were prepared to mimic the microstructural characteristics of AF tissue using electrospinning technique. AF-derived stem cells (AFSCs) were cultured on the PLLA fibrous scaffolds for 7 days. Results: The morphology of AFSCs significantly varied when cultured on the scaffolds with various fiber diameters and orientation. AFSCs were nearly round on scaffolds with small fibers. However, they became spindle-shaped on scaffolds with large fibers. Meanwhile, upregulated expression of collagen-I gene happened in cells cultured on scaffolds with large fibers, while enhanced expression of collagen-II and aggrecan genes was seen on scaffolds with small fibers. The production of related proteins also showed similar trends. Further, culturing AFSCs on a heterogeneous scaffold by overlaying membranes with different fiber sizes led to the formation of a hierarchical structure approximating native AF tissue. Conclusion: Findings from this study demonstrate that fibrous scaffolds with different fiber sizes effectively promoted the differentiation of AFSCs into specific cells similar to the types of cells at various AF zones. It also provides a valuable reference for regulation of cell differentiation and fabrication of engineered tissues with complex hierarchical structures using the physical cues of scaffolds. The translational potential of this article: Effective AF repair is an essential need for treating degenerative disc disease. Tissue engineering is a promising approach to achieving tissue regeneration and restoring normal functions of tissues. By mimicking the key structural features of native AF tissue, including fiber size and alignment, this study deciphered the effect of scaffold materials on the cell differentiation and extracellular matrix deposition, which provides a solid basis for designing new strategies toward more effective AF repair and regeneration.
Article
Full-text available
Recent studies have demonstrated that human astrocytes and fibroblasts can be directly converted into functional neurons by small molecules. However, fibroblasts, as a potentially better cell resource for transplantation, are not as easy to reprogram as astrocytes regarding their fate to neurons, and chemically induced neurons (iNs) with low efficiency from fibroblasts resulted in limited application for the treatment of neurological disorders, including depression. Here, we report that human fibroblasts can be efficiently and directly reprogrammed into glutamatergic neuron-like cells by serially exposing cells to a combination of small molecules. These iNs displayed neuronal transcriptional networks, and also exhibited mature firing patterns and formed functional synapses. Importantly, iNs could integrate into local circuits after transplantation into postnatal mouse brain. Our study provides a rapid and efficient transgene-free approach for chemically generating neuron-like cells from human fibroblasts. Furthermore, our approach offers strategies for disease modeling and drug discovery in central nervous system disorders.
Article
Full-text available
In the past few years, there have been many efforts underway to develop effective wound healing treatments for traumatic injuries. In particular, wound‐healing peptides (WHPs) and peptide‐grafted dressings hold great promise for novel therapeutic strategies for wound management. This study reports a topical formulation of a new synthetic WHP (REGRT, REG) embedded in a hyaluronic acid (HA)‐based hydrogel dressing for the enhancement of acute excisional wound repair. The copper‐free click chemistry is utilized to form biocompatible HA hydrogels by cross‐linking dibenzocyclooctyl‐functionalized HA with 4‐arm poly(ethylene glycol) (PEG) azide. The HA hydrogels are grafted with the REG peptide, a functional derivative of erythroid differentiation regulator1, displaying potent cell motility‐stimulating ability, thus sustainably releasing physiologically active peptides for a prolonged period. Combined with the traditional wound healing benefits of HA, the HA hydrogel embedded REG (REG‐HAgel) accelerates re‐epithelialization in skin wound healing, particularly by promoting migration of fibroblasts, keratinocytes, and endothelial cells. REG‐HAgels improve not only rate, but quality of wound healing with higher collagen deposition and more microvascular formation while being nontoxic. The peptide‐grafted HA hydrogel system can be considered as a promising new wound dressing formulation strategy for the treatment of different types of wounds with combinations of various natural and synthetic WHPs.
Article
Full-text available
Statement of significance: Delivery of RNAi molecules may be a valuable strategy to guide cell behavior for tissue engineering applications, but to date there have been no reports of a biomaterial system capable of both encapsulation of cells and controlled delivery of incorporated RNA. Here, we present PEG hydrogels that form in situ via Michael type reaction, and that permit encapsulation of hMSCs and the concomitant controlled delivery of siNoggin and/or miRNA-20a. These RNAs were chosen to suppress noggin, a BMP-2 antagonist, and/or PPAR-γ, a negative regulator of BMP-2-mediated osteogenesis, and therefore promote osteogenic differentiation of hMSCs and subsequent bone repair in critical-sized rat calvarial defects. Simultaneous delivery of hMSCs and miRNA-20a enhanced repair of these defects compared to hydrogels containing hMSCs without siRNA or with negative control siRNA. This in situ forming PEG hydrogel system offers an exciting platform for healing critical-sized bone defects by localized, controlled delivery of RNAi molecules to encapsulated hMSCs and surrounding cells.
Article
Full-text available
Bone grafting remains the method of choice for the majority of surgeons in the treatment of large bone defects, since it fills spaces and provides support to enhance biological bone repair. Recently, we reported our research on a bioactive multiphase macroporous scaffold with interconnected porous structure, nano-crystal surface microstructure that can release bioactive ions. Moreover, we demonstrated the excellent in vitro biological activity of the scaffold. With this study, we set out to evaluate the in vivo, osteogenesis and vascularization of the scaffold in the treatment of large bone defects (10-mm radial bone defect in rabbits). In comparison with the control group, X-ray and Micro-CT results at the 4th and 8th week post-surgery the bioactive scaffold displayed an enhanced level of new bone and vessel formation. Histological results at the same weeks indicated improved bone formation, osseointegration and new vessel ingrowth inside the bioactive scaffold. These findings establish a good foundation for the potential clinical validation of the bioactive macroporous biomaterial scaffold for use as a bone substitute or in tissue engineering.
Article
Full-text available
Host stem/progenitor cells can be mobilized and recruited to a target location using biomaterials, and these cells may be used for in situ tissue regeneration. The objective of this study was to investigate whether host biologic resources could be used to regenerate renal tissue in situ. Collagen hydrogel was injected into the kidneys of normal mice, and rat kidneys that had sustained ischemia/reperfusion injury. After injection, the kidneys of both animal models were examined up to 4 weeks for host tissue response. The infiltrating host cells present within the injection regions expressed renal stem/progenitor cell markers, PAX-2, CD24, and CD133, as well as mesenchymal stem cell marker, CD44. The regenerated renal structures were identified by immunohistochemistry for renal cell specific markers, including synaptopodin and CD31 for glomeruli and cytokeratin and neprilysin for tubules. Quantitatively, the number of glomeruli found in the injected regions was significantly higher when compared to normal regions of renal cortex. This phenomenon occurred in normal and ischemic injured kidneys. Furthermore, the renal function after ischemia/reperfusion injury was recovered after collagen hydrogel injection. These results demonstrate that introduction of biomaterials into the kidney is able to facilitate the regeneration of glomerular and tubular structures in normal and injured kidneys. Such an approach has the potential to become a simple and effective treatment for patients with renal failure. Stem Cells Translational Medicine 2018;7:241–250
Article
Full-text available
The accumulated evidence points to the microenvironment as the primary mediator of cellular fate determination. Comprised of parenchymal cells, stromal cells, structural extracellular matrix proteins, and signaling molecules, the microenvironment is a complex and synergistic edifice that varies tissue to tissue. Furthermore, it has become increasingly clear that the microenvironment plays crucial roles in the establishment and progression of diseases such as cardiovascular disease, neurodegeneration, cancer, and ageing. Here we review the historical perspectives on the microenvironment, and how it has directed current explorations in tissue engineering. By thoroughly understanding the role of the microenvironment, we can begin to correctly manipulate it to prevent and cure diseases through regenerative medicine techniques.
Article
Full-text available
Clustered regularly interspaced short palindromic repeats (CRISPR)–CRISPR associated protein 9 (Cas9)-based therapeutics, especially those that can correct gene mutations via homology-directed repair, have the potential to revolutionize the treatment of genetic diseases. However, it is challenging to develop homology-directed repair-based therapeutics because they require the simultaneous in vivo delivery of Cas9 protein, guide RNA and donor DNA. Here, we demonstrate that a delivery vehicle composed of gold nanoparticles conjugated to DNA and complexed with cationic endosomal disruptive polymers can deliver Cas9 ribonucleoprotein and donor DNA into a wide variety of cell types and efficiently correct the DNA mutation that causes Duchenne muscular dystrophy in mice via local injection, with minimal off-target DNA damage.
Article
Full-text available
Classical bone tissue engineering involves the use of culture-expanded cells and scaffolds to produce tissue constructs for transplantation. Despite promising results, clinical adoption of these constructs has been limited due to various drawbacks, including extensive cell expansion steps, low cell survival rate upon transplantation, and the possibility of immuno-rejection. To bypass the ex vivo cell culture and transplantation process, we exploited the regenerative capacity of the host by mobilizing endogenous stem cells to the site of injury. Systemic injection of substance P (SP) induced mobilization of CD29⁺ CD105⁺ CD45⁻ cells from bone marrow and enhanced bone tissue regeneration in a critical-sized calvarial bone defect model. To provide an appropriate environment for endogenous stem cells to survive and differentiate into osteogenic lineage cells, electrospun nanofibrous polycaprolactone (PCL) scaffolds were functionalized with hydroxyapatite (HA) particles via a polydopamine (PDA) coating to create highly osteoinductive PCL-PDA-HA scaffolds that were implanted in defects. The combination of the PCL-PDA-HA scaffold and SP treatment enhanced in situ bone tissue formation in defects. Thus, our in situ bone regeneration strategy, which combines recruitment of endogenous stem cells from the bone marrow to defective sites and implantation of a highly biocompatible and osteoinductive cell-free scaffold system, has potential as an effective therapeutic in regenerative medicine.
Article
Full-text available
Our study aims to investigate the effects of the SDF-1/CXCR4 axis on the repair of traumatic brain injury (TBI) in rats by mediating bone marrow derived from mesenchymal stem cells (BMSCs). Healthy male SD rats were collected, their tibiofibulars were removed, cultured, and BMSCs were collected. The expression of cell-surface molecular proteins was examined using flow cytometry. The mRNA and protein expression of CXCR4 in cells were tested using qRT-PCR and western blotting analysis. An electronic brain injury instrument was utilized to build TBI rat models and each rat was assigned into the experiment, positive control and control groups (10 rats in each group). The morris water maze was used to calculate the escape latency and number of times rats in each group crossed the platform. Neurological severity scores (NSS) was calculated to evaluate the recovery of neurological functioning. The distribution of neuronal nuclear antigens was detected using double-labeling immunohistochemistry. The morphological changes in the hippocampal neuronal and the number of BrdU-positive cells were observed through Nissl's staining and high magnification. The mRNA and protein expressions of CXCR4 were gradually increased as SDF-1 concentration increased. NGF and BDNF positive cells were expressed in each group. The distribution of neuronal nuclear antigens in the experiment group was elevated compared to the control and positive control groups. Among the three groups, the experimental group had the shortest escape latency and the highest number platform crossings. The difference in NSS among the three groups was significant. The experimental group had better cell morphology and a higher number of BrdU-positive cells than the other groups. The present study demonstrates that transplanting BMSCs with SDF-1-induced CXCR4 expression can promote the repair of TBI. This is expected to become a new treatment regimen for TBI.
Article
Full-text available
Spinal cord injuries (SCI) often lead to persistent neurological dysfunction due to failure in axon regeneration. Unfortunately, currently established treatments, such as direct drug administration, do not effectively treat SCI due to rapid drug clearance from our bodies. Here, we introduce a three-dimensional aligned nanofibers-hydrogel scaffold as a bio-functionalized platform to provide sustained non-viral delivery of proteins and nucleic acid therapeutics (small non-coding RNAs), along with synergistic contact guidance for nerve injury treatment. A hemi-incision model at cervical level 5 in the rat spinal cord was chosen to evaluate the efficacy of this scaffold design. Specifically, aligned axon regeneration was observed as early as one week post-injury. In addition, no excessive inflammatory response and scar tissue formation was triggered. Taken together, our results demonstrate the potential of our scaffold for neural tissue engineering applications.
Article
Full-text available
Tissue regeneration using stem cell-based transplantation faces many hurdles. Alternatively, therapeutically exploiting endogenous stem cells to regenerate injured or diseased tissue may circumvent these challenges. Here we show resident fibrocartilage stem cells (FCSCs) can be used to regenerate and repair cartilage. We identify FCSCs residing within the superficial zone niche in the temporomandibular joint (TMJ) condyle. A single FCSC spontaneously generates a cartilage anlage, remodels into bone and organizes a haematopoietic microenvironment. Wnt signals deplete the reservoir of FCSCs and cause cartilage degeneration. We also show that intra-articular treatment with the Wnt inhibitor sclerostin sustains the FCSC pool and regenerates cartilage in a TMJ injury model. We demonstrate the promise of exploiting resident FCSCs as a regenerative therapeutic strategy to substitute cell transplantation that could be beneficial for patients suffering from fibrocartilage injury and disease. These data prompt the examination of utilizing this strategy for other musculoskeletal tissues.
Article
Full-text available
The development of cell-free vascular grafts has tremendous potential for tissue engineering. However, thrombus formation, less-than-ideal cell infiltration, and a lack of growth potential limit the application of electrospun scaffolds for in situ tissue-engineered vasculature. To overcome these challenges, here we present development of an acellular tissue-engineered vessel based on electrospun poly(L-lactide-co-e-caprolactone) scaffolds. Heparin was conjugated to suppress thrombogenic responses, and substance P (SP) was immobilized to recruit host cells. SP was released in a sustained manner from scaffolds and recruited human bone marrow-derived mesenchymal stem cells. The biocompatibility and biological performance of the grafts were evaluated by in vivo experiments involving subcutaneous scaffold implantation in Sprague-Dawley rats (n=12) for up to 4 weeks. Histological analysis revealed a higher extent of accumulative host cell infiltration, neotissue formation, collagen deposition, and elastin deposition in scaffolds containing either SP or heparin/SP than in the control groups. We also observed the presence of a large number of laminin-positive blood vessels, von Willebrand factor (vWF+) cells, and alpha smooth muscle actin-positive cells in the explants containing SP and heparin/SP. Additionally, SP and heparin/SP grafts showed the existence of CD90+ and CD105+ MSCs and induced a large number of M2 macrophages to infiltrate the graft wall compared with that observed with the control group. Our cell-free grafts could enhance vascular regeneration by endogenous cell recruitment and by mediating macrophage polarization into the M2 phenotype, suggesting that these constructs may be a promising cell-free graft candidate and are worthy of further in vivo evaluation.
Article
Full-text available
DNA-based gene therapy has considerable therapeutic potential, but the challenges associated with delivery continue to limit progress. Messenger RNA (mRNA) has the potential to provide for transient production of therapeutic proteins, without the need for nuclear delivery and without the risk of insertional mutagenesis. Here we describe the sustained delivery of therapeutic proteins in vivo in both rodents and non-human primates via nanoparticle-formulated mRNA. Nanoparticles formulated with lipids and lipid-like materials were developed for delivery of two separate mRNA transcripts encoding either human erythropoietin (hEPO) or factor IX (hFIX) protein. Dose-dependent protein production was observed for each mRNA construct. Upon delivery of hEPO mRNA in mice, serum EPO protein levels reached several orders of magnitude (>125 000-fold) over normal physiological values. Further, an increase in hematocrit (Hct) was established, demonstrating that the exogenous mRNA-derived protein maintained normal activity. The capacity of producing EPO in non-human primates via delivery of formulated mRNA was also demonstrated as elevated EPO protein levels were observed over a 72-h time course. Exemplifying the possible broad utility of mRNA drugs, therapeutically relevant amounts of human FIX (hFIX) protein were achieved upon a single intravenous dose of hFIX mRNA-loaded lipid nanoparticles in mice. In addition, therapeutic value was established within a hemophilia B (FIX knockout (KO)) mouse model by demonstrating a marked reduction in Hct loss following injury (incision) to FIX KO mice.Gene Therapy advance online publication, 30 June 2016; doi:10.1038/gt.2016.46.
Article
Full-text available
When treating critical limb ischemia caused by various chronic diseases such as diabetes and hypertension, it is essential to effectively induce angiogenesis to supply blood flow to the ischemic region. Recently, several studies have investigated the effects of cell transplantation with mesenchymal stem cells (MSCs) as a therapeutic modality for treating these ischemic diseases. However, some limitations have to be overcome first before cell transplantation can be considered as a promising treatment for ischemic diseases, such as limited sources of cells and the low survival rates of transplanted cells. In this study, self-assembled peptide hydrogels (SAPs) coupled with substance P (SP) were used to induce the recruitment of MSCs to an injury site in mouse ischemic hind limb models without exogenous injection of cells. Additionally, a combined delivery strategy consisting of local and systemic delivery of SP was used to examine the synergetic effects of systemic and local deliveries. Limb ischemia in athymic mice was induced through the femoral artery by ligating and resecting its branches, and SAP coupled with SP (bioactive SAPs) was injected into the ischemic region. The therapeutic effects on the ischemic region were observed in terms of cell migration, fibrosis, apoptosis, and angiogenesis in each experimental group. The combined therapeutic delivery system resulted in the recruitment of more cells for effective regeneration, promotion of neovascularization and formation of mature vessels for tissue perfusion, and inhibition of fibrosis and cell apoptosis compared to a single treatment. In conclusion, it was confirmed that the combined therapy of local and system delivery of the SP conjugated peptide hydrogels and SP could effectively enhance the mobilization of host cells related to angiogenesis to injured tissue, and consequently, they could be useful in treating ischemic diseases without cell transplantation.
Article
Full-text available
The molecular mechanism of bone marrow mesenchymal stromal stem cells (BMSCs) mobilization and migration to the liver was poorly understood. Stromal cell-derived factor-1 (SDF-1) participates in BMSCs homing and migration into injury organs. We try to investigate the role of SDF-1 signaling in BMSCs migration towards injured liver. The expression of CXCR4 in BMSCs at mRNA level and protein level was confirmed by RT-PCR, flow cytometry, and immunocytochemistry. The SDF-1 or liver lysates induced BMSCs migration was detected by transwell inserts. CXCR4 antagonist, AMD3100, and anti-CXCR4 antibody were used to inhibit the migration. The Sprague-Dawley rat liver injury model was established by intraperitoneal injection of thioacetamide. The concentration of SDF-1 increased as modeling time extended, which was determined by ELISA method. The Dir-labeled BMSCs were injected into the liver of the rats through portal vein. The cell migration in the liver was tracked by in vivo imaging system and the fluorescent intensity was measured. In vivo , BMSCs migrated into injured liver which was partially blocked by AMD3100 or anti-CXCR4 antibody. Taken together, the results demonstrated that the migration of BMSCs was regulated by SDF-1/CXCR4 signaling which involved in BMSCs recruitment to injured liver.
Article
Full-text available
In situ tissue regeneration holds great promise for regenerative medicine and tissue engineering applications. However, to achieve control over long-term and localised presence of biomolecules, certain barriers must be overcome. The aim of this study was to develop electrospun scaffolds for the fabrication of artificial vascular grafts that can be remodelled within a host by endogenous cell recruitment. We fabricated scaffolds by mixing appropriate proportions of linear poly (l-lactide-co-ε-caprolactone) (PLCL) and substance P (SP)-immobilised PLCL, using electrospinning to develop vascular grafts. Substance P was released in a sustained fashion from electrospun membranes for up to 30 d, as revealed by enzyme-linked immunosorbent assay. Immobilised SP remained bioactive and recruited human bone marrow-derived mesenchymal stem cells (hMSCs) in an in vitro Trans-well migration assay. The biocompatibility and biological performance of the scaffolds were evaluated by in vivo experiments involving subcutaneous scaffold implantations in Sprague-Dawley rats for up to 28 d followed by histological and immunohistochemical studies. Histological analysis revealed a greater extent of accumulative host cell infiltration and collagen deposition in scaffolds containing higher contents of SP than observed in the control group at both time points. We also observed the presence of a large number of laminin-positive blood vessels and Von Willebrand factor (vWF+) cells in the explants containing SP. Additionally, scaffolds containing SP showed the existence of CD90+ and CD105+ MSCs. Collectively, these findings suggest that the methodology presented here may have broad applications in regenerative medicine, and the novel scaffolding materials can be used for in situ tissue regeneration of soft tissues.
Article
Full-text available
Most organisms experience changes in regenerative abilities through their lifespan. During aging, numerous tissues exhibit a progressive decline in homeostasis and regeneration that results in tissue degeneration, malfunction and pathology. The mechanisms responsible for this decay are both cell intrinsic, such as cellular senescence, as well as cell-extrinsic, such as changes in the regenerative environment. Understanding how these mechanisms impact on regenerative processes is essential to devise therapeutic approaches to improve tissue regeneration and extend healthspan. This review offers an overview of how regenerative abilities change through lifespan in various organisms, the factors that underlie such changes and the avenues for therapeutic intervention. It focuses on established models of mammalian regeneration as well as on models in which regenerative abilities do not decline with age, as these can deliver valuable insights for our understanding of the interplay between regeneration and aging.
Article
Nerve growth factor (NGF) is important for peripheral nerve regeneration. However, its short half‐life and rapid diffusion in body fluids limit its clinical efficacy. Collagen has favorable biocompatibility and biodegradability, but weak immunogenicity. Because it possesses an NGF binding domain, we crosslinked heparin to collagen tubes to construct nerve guidance conduits for delivering NGF. The conduits were implanted to bridge a facial nerve defect in rats. Histological and functional analyses were performed to assess the effect of the nerve guidance conduit on facial nerve regeneration. Heparin enhanced the binding of NGF to collagen while retaining its bioactivity. Also, the nerve guidance conduit significantly promoted axonal growth and Schwan cell proliferation at 12 weeks after surgery. The nerve regeneration and functional recovery outcomes using the nerve guidance conduit were similar to those of autologous nerve grafting. Therefore, the nerve guidance conduit may promote safer nerve regeneration.
Article
In situ tissue regeneration harnesses the body’s regenerative potential to control cell functions for tissue repair. The design of biomaterials for in situ tissue engineering requires precise control over biophysical and biochemical cues to direct endogenous cells to the site of injury. These cues are required to induce regeneration by modulating the extracellular microenvironment or driving cellular reprogramming. In this Review, we outline two biomaterials approaches to control the regenerative capacity of the body for tissue-specific regeneration. The first approach includes the use of bioresponsive materials with an ability to direct endogenous cells, including immune cells and progenitor or stem cells, to facilitate tissue healing, integration and regeneration. The second approach focuses on in situ cellular reprogramming via delivery of transcription factors, RNA-based therapeutics, in vivo gene editing and biomaterials-driven epigenetic transformation. In addition, we highlight tools for engineering the next generation of biomaterials to modulate in situ tissue regeneration. Overall, leveraging the regenerative potential of the human body via engineered biomaterials is a simple and effective approach to replace injured or diseased tissues. In situ tissue regeneration harnesses the body’s regenerative potential for tissue repair using engineered biomaterials. In this Review, we outline various biomaterials approaches to control the body’s regenerative capacity for tissue-specific regeneration by modulating the extracellular microenvironment or driving cellular reprogramming.
Article
Biomaterial topography-based strategy is regarded as an effective way to regulate osteoimmune environment which plays an indispensable role in bone regeneration process. The rapid development of manufacture techniques makes it possible to investigate the cell-topography interactions by preparing various micro and nano-topographical surfaces on biomaterials. Still, it is a challenge to prepare well-defined micro/nano hierarchical structures of bioceramics due to the inherent brittleness of ceramic materials. Also, the correlation between osteoimmunomodulation initiated by micro/nano hierarchical topographies and the tissue regeneration outcomes is unclear. In this study, we prepared well-defined micro/nano hierarchical structures on hydroxyapatite (HA) bioceramics through the combination of the photolithography and hydrothermal techniques. Three different microscale circular patterns (4 ?m, 12 ?m and 36 ?m) and nanotopographies (nanoneedle, nanosheet and nanorod) were fabricated by changing the size of mask and the condition of the hydrothermal reaction. The macrophage responses on the nanoneedle structures with different micropatterns were investigated and the micro/nano hierarchical structures with appropriate pattern size could either promote or alleviate the inflammation, which further affected the outcomes of the osteogenic differentiation of human bone marrow stromal cells (hBMSCs) and angiogenic acticvity of human umbilical vein endothelial cells (HUVECs). Our study demonstrated that the osteoimmunomodulation could be manipulated via tuning the micro/nano hierarchical structures, which could lead to a new strategy for the development of bone biomaterials with favorable osteoimmunomodulatory properties.
Article
The nature of the protein corona forming on a biomaterial surfaces can affect the performance of implanted devices. This study investigated the role of surface chemistry and wettability on human serum-derived protein corona formation on biomaterial surfaces, and the subsequent effects on cellular innate immune response. Plasma polymerization, a substrate independent technique, was employed to create nano-thin coatings with four specific chemical functionalities, and a spectrum of surface charges and wettability. The amount and type of protein adsorbed was strongly influenced by surface chemistry and wettability, but did not show any dependence on surface charge. An enhanced adsorption of the dysopsonin albumin was observed on hydrophilic carboxyl surfaces while high opsonin IgG2 adsorption was seen on hydrophobic hydrocarbon surfaces. This in turn led to distinct immune response from macrophages; hydrophilic surfaces drove greater expression of anti-inflammatory cytokines by macrophages, whilst surface hydrophobicity caused increased production of pro-inflammatory signaling molecules. These findings map out a unique relationship between surface chemistry, hydrophobicity, protein corona formation and subsequent cellular innate immune responses; the potential outcomes of these studies may be employed to tailor biomaterial surface modifications, to modulate serum protein adsorption and to achieve desirable innate immune response to implanted biomaterials and devices.
Article
mRNA therapeutics have recently experienced a new wave of interest, mainly due to the discovery that chemical modifications to mRNA's molecular structure could drastically reduce its inherent immunogenicity and perceived instability. On this basis, we aimed to explore the potential of chemically stabilized mRNA for ocular applications. More specifically, we investigated the behavior of mRNA-loaded lipid-based carriers in human retinal cells (in vitro), in bovine retinal explants (ex vivo) and in mouse retinas (in vivo). We demonstrate a clear superiority of mRNA over pDNA to induce protein expression in different retinal cell types, which was further enhanced by chemical modification of the mRNA, providing up to ~1800-fold higher reporter gene expression compared to pDNA. Moreover, transgene expression could be detected for at least 20 days after a single administration of chemically modified mRNA in vitro. We furthermore determined the localization and extent of mRNA expression depending on the administration route. After subretinal (SR) administration, mRNA expression was observed in vivo and ex vivo. By contrast, intravitreal (IVT) administration resulted in limited expression in vivo. Using ex vivo bovine explants with an intact vitreoretinal (VR) interface we could attribute this to the inner limiting membrane (ILM), which presents a large barrier for non-viral delivery of mRNA, trapping mRNA complexes at the vitreal side. When the vitreous was removed, which compromises the ILM, mRNA expression was apparent and seemed to colocalize with Müller cells or photoreceptors after respectively IVT or SR administration. Taken together, this study represents a first step towards mRNA-mediated therapy for retinal diseases.
Article
Recently, we found that although high-stiffness matrices stimulated osteogenic differentiation of bone marrow-derived stromal cells (BMSCs), the macrophages (Mφs) in high-stiffness transglutaminase crosslinked gelatins (TG-gels) tended to undergo M1 polarization and hence compromised cell osteogenesis. In this study, we hypothesized that the copresentation of interleukin (IL)-4 and stromal cell-derived factor (SDF)-1α in high-stiffness TG-gels may enhance periodontal regeneration by modulating Mφ polarization and promoting endogenous stem cell recruitment. We found that Mφs were more likely to polarize toward an immunomodulatory M2 state in the presence of IL-4 and hence positively influence the osteogenic differentiation of BMSCs when these cells coexisted in either indirect or direct co-culture systems. In cell migration assays, BMSCs exhibited an enhanced capability to move toward gels containing SDF-1α, and more cells could be recruited into the three-dimensional matrix of TG-gels. When TG-gels containing IL-4 and/or SDF-1α were used to repair periodontal defects, more new bone (MicroCT) was formed in animals that received the dual cytokine-loaded transplants at 4 weeks postsurgery. Mφs were recruited to all the transplanted gels, and after one week, more M1-phenotype cells were found in the groups without IL-4, while the presence of IL-4 was more likely to result in M2 polarization (immunofluorescence staining). When the tissue biopsies were histologically examined, the TG-gels containing both IL-4 and SDF-1α led to a generally satisfactory regeneration with respect to attachment recovery (epithelial and connective tissue) and hybrid tissue regeneration (bone, periodontal ligament and cementum). Our data suggest that the incorporation of IL-4 into high-stiffness TG-gels may promote the M2 polarization of Mφs and that SDF-1α can be applied to guide endogenous cell homing. Overall, building capacity for Mφ modulation and cell recruitment in high-stiffness hydrogels represents a simple and effective strategy that can support high levels of periodontal tissue regeneration. Statement of significance: The development of hydrogel-based regenerative therapies centered on the mobilization and stimulation of native cells for therapeutics opens a window toward realizing periodontal endogenous regeneration. In the present study, the parallel use of immunomodulatory and homing factors in high-stiffness hydrogel materials is shown to induce stem cell homing, modulate cell differentiation and indeed induce regrowth of the periodontium. We found that incorporation of interleukin (IL)-4 in high-stiffness TG-gels coaxed macrophages to polarize into M2 phenotypes, and stromal cell-derived factor (SDF)-1α could be applied to direct endogenous cell homing. Hence, we present for the first time a clinically relevant strategy based on macrophage modulation and host cell recruitment that can support high levels of periodontal tissue regeneration.
Article
Bovine xenograft materials, followed by synthetic biomaterials, which unfortunately still lack documented predictability and clinical performance, dominate the market for the cranio‐maxillofacial area. In Europe, new stringent regulations are expected to further limit the allograft market in the future Aim Within this narrative review, we discuss possible future biomaterials for bone replacement. Scientific Rationale for Study Although the bone graft (BG) literature is overflooded, only a handful of new BG substitutes are clinically available. Laboratory studies tend to focus on advanced production methods and novel biomaterial features, which can be costly to produce. Practical Implications In this review, we ask why such a limited number of BGs are clinically available when compared to extensive laboratory studies. We also discuss what features are needed for an ideal BG. Results We have identified the key properties of current bone substitutes and have provided important information to guide clinical decision‐making and generate new perspectives on bone substitutes. Our results indicated that different mechanical and biological properties are needed despite each having a broad spectrum of variations. Conclusions We foresee bone replacement composite materials with higher levels of bioactivity, providing an appropriate balance between bioabsorption and volume maintenance for achieving ideal bone remodelling. This article is protected by copyright. All rights reserved.
Article
The design of tissue engineered scaffolds based on polymerized high internal phase emulsions (polyHIPEs) has emerged as a promising bone grafting strategy. We previously reported the ability to 3D print emulsion inks to better mimic the structure and mechanical properties of native bone while precisely matching defect geometry. In the current study, redox-initiated hydrogel carriers were investigated for in situ delivery of human mesenchymal stem cells (hMSCs) utilizing the biodegradable macromer, poly(ethylene glycol)-dithiothreitol. Hydrogel carrier properties including network formation time, sol-gel fraction, and swelling ratio were modulated to achieve rapid cure without external stimuli and a target cell-release period of 5-7 days. These in situ carriers enabled improved distribution of hMSCs in 3D printed polyHIPE grafts over standard suspension seeding. Additionally, carrier-loaded polyHIPEs supported sustained cell viability and osteogenic differentiation of hMSCs post-release. In summary, these findings demonstrate the potential of this in situ curing hydrogel carrier to enhance the cell distribution and retention of hMSCs in bone grafts. Although initially focused on improving bone regeneration, the ability to encapsulate cells in a hydrogel carrier without relying on external stimuli that can be attenuated in large grafts or tissues is expected to have a wide range of applications in tissue engineering.
Article
Application of stem cells in combination with nanofibrous substrates is an interesting biomimetic approach for enhanced regeneration of damaged tissues such as bone and cartilage. The investigation of the complex interplay between nanotopographical cues of niche and noncoding RNAs in stem cells fate is an effective tool to find a new strategy for enhancing the induction of osteogenesis. In this study, we investigated the effects of aligned and random orientations of nanofibers as a natural ECM-mimicking environment on the network of noncoding RNA in mesenchymal stem cells. Aligned and randomly oriented Ploy (L-lactide) PLLA scaffolds were fabricated via electrospinning. Human Adipose Tissue-Derived Mesenchymal Stem Cells (hASCs) were isolated from adipose tissue and were cultured on surfaces of these scaffolds. Their capacity to support hMSCs proliferation was also investigated by MTT assay and the expression of c-Myc gene. Then, after 7, 14 and 21 days, the osteogenic commitment of hMSCs and the miRNA regulatory network in BMP signaling pathway were evaluated by measuring alkaline phosphatase (ALP) activity, extracellular calcium deposition, and bone-related gene activation by Real-Time PCR. Furthermore, osteogenic differentiation was evaluated with regard to their noncoding RNA network. Our results for the first time showed an interaction between nanotopographical cues and miRNA activity in hMSCs. We found that the nanotopographical cues could be used to influence the osteogenic differentiation process of hMSCs through the modulation of lncRNAs and miR-125b as negative regulators of osteogenesis as well as the H19 modulator BMP signaling pathway that acts as a miRNA sponge. Moreover, we also demonstrated for the first time that MEG3 as a long noncoding RNA is controlled by miR-125b and microRNA-triggered lncRNA decay mechanism. This strategy seems to be an important tool for controlling stem cell fate in engineered tissues and provide new insights into most biocompatible scaffolds for bone-graft substitutes.
Article
In contrast to non-mammalian vertebrates, mammals and humans have limited innate capacity for the self-regeneration of tissues and organs owing to differences in genetics, development, immune systems and tissue complexity. Endogenous stem cells are tissue-specific adult stem cells with the capacity to self-renew and differentiate into specific cell types. Therefore, endogenous stem cells are being explored for the regeneration of tissues in situ and in vivo. Stem cells reside in specific niches in the body, and stem cell activation depends on progressive changes in the niche. Niches are specific and instructive microenvironments that can be recreated using biomaterial-based scaffolds. Such scaffolds can be fabricated into a variety of shapes and formulations, and they can be functionalized with biochemical and biophysical cues to guide stem cell fate and migration. In this Review, we discuss important differences in the self-regeneration abilities of non-mammalian vertebrates and mammals, including humans, and investigate adult stem cell populations and their niches involved in tissue repair and regeneration. We highlight natural and synthetic biomaterials and their potential for improving applications of endogenous stem cells and examine the role of interspecies chimaeras in regenerative medicine.
Article
Human stem cells hold significant potential for the treatment of various diseases. However, their use as a therapy is hampered because of limited understanding of the mechanisms by which they respond to environmental stimuli. Efforts to understand extracellular biophysical cues have demonstrated the critical roles of geometrical and mechanical signals in determining the fate of stem cells. The goal of this study was to explore the interplay between cell polarity and matrix stiffness in stem cell lineage specification. We hypothesize that confining cells to asymmetric extracellular matrix islands will impart polarity at a single-cell level and will interact with mechanical signals to define the lineage of stem cells. To test these hypotheses, we employed microcontact printing to create patterned symmetric and asymmetric hydrogel islands of soft and hard surface stiffness. Human mesenchymal stem cells (hMSCs) were confined to these islands at the single-cell level and given the ability to differentiate along adipogenic or osteogenic routes. Our results demonstrated that cell polarity defines the lineage specification of hMSCs only on islands with low stiffness. Insight gained from this study provides a rational basis for designing stem cell cultures to enhance tissue engineering and regenerative medicine strategies.
Article
Statement of significance: The substrate stiffness of a scaffold plays critical roles in modulating both reparative cells, such as mesenchymal stem cells (MSCs), and immune cells, such as macrophages (Mφs). Although the influences of material stiffness on either Mφs or MSCs, have been extensively described, how the two cell types respond to matrix cues to dynamically affect each other in a three-dimensional (3D) biosystem remains largely unknown. Here, we report our findings that, in a platform wherein Mφs and bone marrow-derived MSCs coexist, matrix stiffness can influence stem cell fate through both direct matrix-associated regulation and indirect Mφ-based modulation. Our data support future studies of the MSC-Mφ-matrix interplay in the 3D context to optimize matrix parameters for the development of the next biomaterial.
Article
The impact of additive manufacturing in our lives has been increasing constantly. One of the frontiers in this change is the medical devices. 3D printing technologies not only enable the personalization of implantable devices with respect to patient-specific anatomy, pathology and biomechanical properties but they also provide new opportunities in related areas such as surgical education, minimally invasive diagnosis, medical research and disease models. In this review, we cover the recent clinical applications of 3D printing with a particular focus on implantable devices. The current technical bottlenecks in 3D printing in view of the needs in clinical applications are explained and recent advances to overcome these challenges are presented. 3D printing with cells (bioprinting); an exciting subfield of 3D printing, is covered in the context of tissue engineering and regenerative medicine and current developments in bioinks are discussed. Also emerging applications of bioprinting beyond health, such as biorobotics and soft robotics, are introduced. As the technical challenges related to printing rate, precision and cost are steadily being solved, it can be envisioned that 3D printers will become common on-site instruments in medical practice with the possibility of custom-made, on-demand implants and, eventually, tissue engineered organs with active parts developed with biorobotics techniques.
Article
Statement of significance: Electrospun aligned fibers, resembling the ultrastructure of tendon, have been previously reported to promote tenogenesis. However, the underlying mechanism is unclear and the aligned fibers alone are not capable enough to commit teno-differentiation of stem cells. The uniqueness of our studies are as follows, based on our observation of reduced expression of histone deacetylases (HDACs) in tendon stem/progenitor cells (TSPCs) cultured on aligned fibers, we proposed a strategy to enhance the tenogenesis effect of aligned fibers by using a small molecule Trichostatin A (TSA), a HDAC inhibitor. Such a TSA-laden poly (L-lactic acid) (PLLA) aligned fiber (A-TSA) scaffold was successfully fabricated by a stable jet electrospinning method, and demonstrated its sustained capability in releasing TSA. The incorporation and subsequent release of bioactive small molecule TSA into electrospun aligned fibers allows a controllable manner for both biochemical and physical regulation of tenogenesis of stem cells both in vitro and in vivo. Collectively, the present study provides a model of "translating the biological knowledge learned from cell-material interaction into optimizing biomaterials (from Biomat-to-Biomat)".
Article
Statement of significance: Silicate bioceramics have been widely used for orthopeadic tissue regeneration because of their excellent characteristics in bone formation. However, there are few studies concerning their interrelationships with the host immune defense that has been proved to greatly influence osteogenesis. In our present study, the akermanite and nagelschmidtite were used as two representative silicate bioceramics to investigate the inflammation responses in vitro and in vivo; and for the first time, the bioactive ions released from the silicate bioceramics were discovered to regulate the macrophage immune responses through both inhibiting the inflammatory signaling and activating apoptosis of macrophages. Our findings in this study may not only increase the understanding in osteogenic activity of silicate bioceramics, but also provide profitable guidance for designing and manufacturing new biomaterials for bone tissue engineering.
Article
Tissue repair after injury is a complex, metabolically demanding process. Depending on the tissue's regenerative capacity and the quality of the inflammatory response, the outcome is generally imperfect, with some degree of fibrosis, which is defined by aberrant accumulation of collagenous connective tissue. Inflammatory cells multitask at the wound site by facilitating wound debridement and producing chemokines, metabolites, and growth factors. If this well-orchestrated response becomes dysregulated, the wound can become chronic or progressively fibrotic, with both outcomes impairing tissue function, which can ultimately lead to organ failure and death. Here we review the current understanding of the role of inflammation and cell metabolism in tissue-regenerative responses, highlight emerging concepts that may expand therapeutic perspectives, and briefly discuss where important knowledge gaps remain. Copyright 2016 by the American Association for the Advancement of Science; all rights reserved.
Article
Tissue engineered scaffolds have emerged as a promising solution for heart valve replacement because of their potential for regeneration. However, traditional heart valve tissue engineering has relied on resource-intensive, cell-based manufacturing, which increases cost and hinders clinical translation. To overcome these limitations, in situ tissue engineering approaches aim to develop scaffold materials and manufacturing processes that elicit endogenous tissue remodeling and repair. Yet despite recent advances in synthetic materials manufacturing, there remains a lack of cell-free, automated approaches for rapidly producing biomimetic heart valve scaffolds. Here, we designed a jet spinning process for the rapid and automated fabrication of fibrous heart valve scaffolds. The composition, multiscale architecture, and mechanical properties of the scaffolds were tailored to mimic that of the native leaflet fibrosa and assembled into three dimensional, semilunar valve structures. We demonstrated controlled modulation of these scaffold parameters and show initial biocompatibility and functionality in vitro. Valves were minimally-invasively deployed via transapical access to the pulmonary valve position in an ovine model and shown to be functional for 15 h.
Article
All cells sense and integrate mechanical and biochemical cues from their environment to orchestrate organismal development and maintain tissue homeostasis. Mechanotransduction is the evolutionarily conserved process whereby mechanical force is translated into biochemical signals that can influence cell differentiation, survival, proliferation and migration to change tissue behavior. Not surprisingly, disease develops if these mechanical cues are abnormal or are misinterpreted by the cells - for example, when interstitial pressure or compression force aberrantly increases, or the extracellular matrix (ECM) abnormally stiffens. Disease might also develop if the ability of cells to regulate their contractility becomes corrupted. Consistently, disease states, such as cardiovascular disease, fibrosis and cancer, are characterized by dramatic changes in cell and tissue mechanics, and dysregulation of forces at the cell and tissue level can activate mechanosignaling to compromise tissue integrity and function, and promote disease progression. In this Commentary, we discuss the impact of cell and tissue mechanics on tissue homeostasis and disease, focusing on their role in brain development, homeostasis and neural degeneration, as well as in brain cancer.
Article
In this study, a cell-free bone tissue engineering system based on a silk fibroin (SF)/nano-hydroxyapatite (nHAp) scaffold was developed, in which two bioactive molecules, stromal cell derived factor-1 (SDF-1) and bone morphogenetic protein-2 (BMP-2), were embedded and released in a sequential and controlled manner to facilitate cell recruitment and bone formation, respectively. BMP-2 was initially loaded into SF microspheres, and these BMP-2 containing microspheres were subsequently encapsulated into the SF/nHAp scaffolds, which were successively functionalized with SDF-1 via physical adsorption. The results indicated rapid initial release of SDF-1 during the first few days, followed by slow and sustained release of BMP-2 for as long as three weeks. The composite scaffold significantly promoted the recruitment of bone marrow mesenchymal stem cells (BMSCs) and osteogenic differentiation of them in vitro. Further, the in vivo studies using D-Luciferin-labeled BMSCs indicated that implantation of this composite scaffold markedly promoted the recruitment of BMSCs to the implanted sites. Enhanced bone regeneration was identified at 12 weeks’ post-implantation. Taken together, our findings suggested that the sequential and sustained release of SDF-1 and BMP-2 from the SF/nHAp scaffolds resulted in a synergistic effect on bone regeneration. Such a composite system, therefore, shows promising potential for cell-free bone tissue engineering applications.
Article
Statement of significance: Organic coatings have been proposed as a solution to foster osseointegration of orthopedic implants. Among them, extracellular matrix-derived peptide motifs are an interesting biomimetic strategy to harness cell-surface interactions. Nonetheless, the combination of multiple peptide motifs in a controlled manner is essential to achieve receptor specificity and fully exploit the potentiality of synthetic peptides. Herein, we covalently graft to titanium a double branched molecule to guide stem cell fate in vitro and generate an osseoinductive titanium surface in vivo. Such synthetic ligand allows for the simultaneous presentation of two bioactive motifs, thus is ideal to test the effect of synergic sequences, such as RGD and PHSRN, and is a clear example of the versatility and feasibility of rationally designed biomolecules.
Article
A bioactive scaffold with desired microstructure is of great importance to induce infiltration of somatic and stem cells, and thereby to achieve the in situ inductive tissue regeneration. In this study, a scaffold with oriented pores in the radial direction is prepared by using methacrylated hyaluronic acid (HA-MA) via controlled directional cooling of a HA-MA solution, and followed with photo-crosslinking to stabilize the structure. Poly(lactide-co-glycolide) (PLGA) is further infiltrated to enhance the mechanical strength, resulting in a compressive modulus of 120 kPa. In vitro culture of bone marrow stem cells (BMSCs) reveals spontaneous cell aggregation inside this type of scaffold with a spherical morphology. In vivo transplantation of the cell-free scaffold in rabbit knees for 12 w regenerates simultaneously both cartilage and subchondral bone with a Wakitani score of 2.8. Moreover, the expression of inflammatory factor interleukin-1β (IL-1β) is down regulated, although tumor necrosis factor-α (TNF-α) is remarkably up regulated. With the anti-inflammatory, bioactive properties and good restoration of full thickness cartilage defect in vivo, the oriented macroporous HA-MA/PLGA hybrid scaffold has a great potential for the practical application in the in situ cartilage regeneration.
Article
Alzheimer's disease (AD) pathogenesis is considered to be the metabolic imbalance between anabolism and clearance of amyloid-beta (Aβ), and the strategy of breaking the equilibrium between soluble and insoluble forms of Aβ is likely to help prevent the progression of AD. Neprilysin (NEP) plays a major role in the clearance of Aβ in the brain, and its supplementation using viral vectors has shown to decrease Aβ deposition and prevent pathogenic changes in the brain. In this study, we developed a new therapeutic strategy by mRNA-based gene introduction. mRNA has the advantages of negligible risk of random integration into genome and not needing to be transcribed precludes the need for nuclear entry. This allows efficient protein expression in slowly-dividing or non-dividing cells, such as neural cells. We constructed mRNA encoding the mouse NEP protein and evaluated its ability degrade Aβ. In vitro transfection of NEP mRNA to primary neurons exhibited Amyloid Precursor Protein (APP) degradation activity superior to that of NEP encoding plasmid DNA. We then evaluated the in vivo activity of NEP mRNA by intracerebroventricular (i.c.v.) infusion using a cationic polymer-based PEGylated nanocarrier to form polyplex nanomicelles, which have been shown to have a high potential to deliver mRNA to various target tissues and organs. Nanomicelles carrying a GFP-NEP fusion mRNA produced efficient protein expression in a diffuse manner surrounding the ventricular space. An ELISA evaluation revealed that the mRNA infusion significantly augmented NEP level and effectively reduced the concentration of Aβ that had been supplemented in the mouse brain. To the best of our knowledge, this is the first study to demonstrate the therapeutic potential of introducing exogenous mRNA for the treatment of brain diseases, opening the new era of mRNA-based therapeutics.
Article
In this study, the wound closure of mouse skin defects was examined in terms of recruitment of mesenchymal stem cells (MSC) and macrophages. For the cells recruitment, stromal derived factor-1 (SDF-1) of a MSC recruitment agent and sphingosine-1 phosphate agonist (SEW2871) of a macrophages recruitment agent were incorporated into gelatin hydrogels, and then released in a controlled fashion. When applied to a skin wound defect of mice, gelatin hydrogels incorporating mixed 500 ng SDF-1 and 0.4, 0.8, or 1.6 mg SEW2871-micelles recruited a higher number of both MSC and macrophages than those incorporating SDF-1 or phosphate buffered saline. However, the number of M1 phenotype macrophages for the hydrogel incorporating mixed SDF-1 and SEW2871-micelles recruited was remarkably low to a significant extent compared with that for those hydrogel incorporating 0.4, 0.8, or 1.6 mg SEW2871-micelles. On the other hand, the number of M2 macrophages 3 days after the implantation of the hydrogels incorporating SDF-1 and 0.4 mg SEW2871-micelles significantly increased compared with that for other hydrogels. In vivo experiments revealed the hydrogels incorporating SDF-1 and 0.4 mg SEW2871-micelles promoted the wound closure of skin defect to a significant stronger extent than those incorporating SEW2871-micelles, SDF-1, and a mixture of SDF-1 and higher doses of SEW2871-micelles. It is concluded that the in vivo recruitment of MSC and macrophages to the defects may contribute to the tissue regeneration of skin wound. This article is protected by copyright. All rights reserved.
Article
Stem cell therapy and tissue engineering hold considerable potential for innovative and transformative strategies to repair damaged tissue form and function. Although many approaches are adopting ex vivo expanded cells for transplantation, an alternative is to manipulate the biomaterial–host interactions that recruit the patients' own stem cells endogenously for regeneration. There are several considerations in targeting the biomaterial–host interactions therapeutically, not the least of which is the biomimetic design of extracellular matrix (ECM)-mimicking materials and the administration of navigation cues and small molecules that target specific aspects of the native healing cascades to stimulate homing of endogenous stem cells and, thereafter, their expansion and differentiation. A sequence of coordinated interactions between the local niche cells and implanted biomaterials offers signals and sign posts that may instruct the cells traveling toward the injured tissues. Furthermore, stem cell function is critically influenced by extrinsic signals provided by the niche as well as by the implanted biomaterials. Novel strategies harnessing growth factors and immunological cues to design materials not only can modulate the behavior of stem cells but also can alter innate and adaptive immunity in a controlled manner. We envisage that successful and safe endogenous regeneration will involve at least three aspects, i.e., homing of sufficient stem cells, controlling cell fate determination, and blunting host immune responses to outside biomaterial devices. Improving our understanding of the biological and physicochemical signals of biomimetic biomaterials that govern immunomodulation for in situ tissue regeneration, particularly context-dependent macrophage (Mφ) polarization, will lead to a concurrent improvement in clinical outcomes.
Article
Vascularization of bone defects is considered a crucial component to the successful regeneration of large bone defects. Although vascular endothelial growth factor (VEGF) has been delivered to critical-size bone defect models to augment blood vessel infiltration into the defect area, its potential to increase bone repair remains ambiguous. In this study, we investigated whether integrin-specific biomaterials modulate the effects of VEGF on bone regeneration. We engineered protease-degradable, VEGF-loaded polyethylene glycol (PEG) hydrogels functionalized with either a triple-helical, α2 β1 integrin-specific peptide (GFOGER) or an αv β3 integrin-targeting peptide (RGD). Covalent incorporation of VEGF into the PEG hydrogel allowed for protease degradation-dependent release of the protein while maintaining VEGF bioactivity. When applied to critical-size segmental defects in the murine radius, GFOGER-functionalized VEGF-free hydrogels exhibited significantly increased vascular volume and density and resulted in a larger number of thicker blood vessels compared to RGD-functionalized VEGF-free hydrogels. VEGF-loaded RGD hydrogels increased vascularization compared to VEGF-free RGD hydrogels, but the levels of vascularization for these VEGF-containing RGD hydrogels were similar to those of VEGF-free GFOGER hydrogels. VEGF transiently increased bone regeneration in RGD hydrogels but had no effect at later time points. In GFOGER hydrogels, VEGF did not show an effect on bone regeneration. However, VEGF-free GFOGER hydrogels resulted in increased bone regeneration compared to VEGF-free RGD hydrogels. These findings demonstrate the importance of integrin-specificity in engineering constructs for vascularization and associated bone regeneration. This article is protected by copyright. All rights reserved.
Article
Extracellular matrix (ECM) is a non-cellular three-dimensional macromolecular network composed of collagens, proteoglycans/glycosaminoglycans, elastin, fibronectin, laminins, and several other glycoproteins. Matrix components bind each other as well as cell to adhesion receptors forming a complex network into which cells reside in all tissues and organs. Cell surface receptors transduce signals into cells from ECM, which regulate diverse cellular functions, such as survival, growth, migration and differentiation and are vital for maintaining normal homeostasis. ECM is a highly dynamic structural network that continuously undergoes remodeling mediated by several matrix-degrading enzymes during normal and pathological conditions. Deregulation of ECM composition and structure is associated with the development and progression of several pathologic conditions. This article emphasizes in the complex ECM structure as to provide a better understanding of its dynamic structural and functional multipotency. Where relevant, the implication of the various families of ECM macromolecules in health and disease is also presented.