scholarly journals The Regeneration of Large-Sized and Vascularized Adipose Tissue Using a Tailored Elastic Scaffold and dECM Hydrogels

2021 ◽  
Vol 22 (22) ◽  
pp. 12560
Author(s):  
Su Hee Kim ◽  
Donghak Kim ◽  
Misun Cha ◽  
Soo Hyun Kim ◽  
Youngmee Jung

A dome-shaped elastic poly (l-lactide-co-caprolactone) (PLCL) scaffold with a channel and pore structure was fabricated by a combinative method of 3D printing technology and the gel pressing method (13 mm in diameter and 6.5 mm in thickness) for patient-specific regeneration. The PLCL scaffold was combined with adipose decellularized extracellular matrix (adECM) and heart decellularized extracellular matrix (hdECM) hydrogels and human adipose-derived stem cells (hADSCs) to promote adipogenesis and angiogenesis. These scaffolds had mechanical properties similar to those of native adipose tissue for improved tissue regeneration. The results of the in vitro real-time PCR showed that the dECM hydrogel mixture induces adipogenesis. In addition, the in vivo study at 12 weeks demonstrated that the tissue-engineered PLCL scaffolds containing the hydrogel mixture (hdECM/adECM (80:20)) and hADSCs promoted angiogenesis and adipose tissue formation, and suppressed apoptosis. Therefore, we expect that our constructs will be clinically applicable as material for the regeneration of patient-specific large-sized adipose tissue.

2020 ◽  
Vol 10 (3) ◽  
pp. 66
Author(s):  
Kateryna Yatsenko ◽  
Iryna Lushnikova ◽  
Alina Ustymenko ◽  
Maryna Patseva ◽  
Iryna Govbakh ◽  
...  

Brain inflammation is a key event triggering the pathological process associated with many neurodegenerative diseases. Current personalized medicine and translational research in neurodegenerative diseases focus on adipose-derived stem cells (ASCs), because they are patient-specific, thereby reducing the risk of immune rejection. ASCs have been shown to exert a therapeutic effect following transplantation in animal models of neuroinflammation. However, the mechanisms by which transplanted ASCs promote cell survival and/or functional recovery are not fully understood. We investigated the effects of ASCs in in vivo and in vitro lipopolysaccharide (LPS)-induced neuroinflammatory models. Brain damage was evaluated immunohistochemically using specific antibody markers of microglia, astroglia and oligodendrocytes. ASCs were used for intracerebral transplantation, as well as for non-contact co-culture with brain slices. In both in vivo and in vitro models, we found that LPS caused micro- and astroglial activation and oligodendrocyte degradation, whereas the presence of ASCs significantly reduced the damaging effects. It should be noted that the observed ASCs protection in a non-contact co-culture suggested that this effect was due to humoral factors via ASC-released biomodulatory molecules. However, further clinical studies are required to establish the therapeutic mechanisms of ASCs, and optimize their use as a part of a personalized medicine strategy.


Author(s):  
Svenja Nellinger ◽  
Ivana Mrsic ◽  
Silke Keller ◽  
Simon Heine ◽  
Alexander Southan ◽  
...  

Due to its availability and minimal invasive harvesting human adipose tissue-derived extracellular matrix (dECM) is often used as a biomaterial in various tissue engineering and healthcare applications. Next to dECM, cell-derived ECM (cdECM) can be generated by and isolated from in vitro cultured cells. So far both types of ECM were investigated extensively towards their application as (bio)material in tissue engineering and healthcare. However, a systematic characterization and comparison of soft tissue dECM and cdECM is still missing. In this study, we characterized dECM from human adipose tissue, as well as cdECM from human adipose-derived stem cells (ASCs), towards their molecular composition, structural characteristics, and biological purity. The dECM was found to exhibit higher levels of collagens and lower levels of sulfated glycosaminoglycans (sGAGs) compared to cdECMs. Structural characteristics revealed an immature state of the fibrous part of cdECM samples. By the identified differences, we aim to support researchers in the selection of a suitable ECM-based biomaterial for their specific application and the interpretation of obtained results.


2019 ◽  
Vol 10 (1) ◽  
Author(s):  
Meifeng Zhu ◽  
Wen Li ◽  
Xianhao Dong ◽  
Xingyu Yuan ◽  
Adam C. Midgley ◽  
...  

Abstract Implanted scaffolds with inductive niches can facilitate the recruitment and differentiation of host cells, thereby enhancing endogenous tissue regeneration. Extracellular matrix (ECM) scaffolds derived from cultured cells or natural tissues exhibit superior biocompatibility and trigger favourable immune responses. However, the lack of hierarchical porous structure fails to provide cells with guidance cues for directional migration and spatial organization, and consequently limit the morpho-functional integration for oriented tissues. Here, we engineer ECM scaffolds with parallel microchannels (ECM-C) by subcutaneous implantation of sacrificial templates, followed by template removal and decellularization. The advantages of such ECM-C scaffolds are evidenced by close regulation of in vitro cell activities, and enhanced cell infiltration and vascularization upon in vivo implantation. We demonstrate the versatility and flexibility of these scaffolds by regenerating vascularized and innervated neo-muscle, vascularized neo-nerve and pulsatile neo-artery with functional integration. This strategy has potential to yield inducible biomaterials with applications across tissue engineering and regenerative medicine.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2400-2400
Author(s):  
Renata Giardini Rosa ◽  
Juares E. Romero Bianco ◽  
Gabriela Pereira dos Santos ◽  
Stephen D. Waldman ◽  
Joanna Weber ◽  
...  

Abstract Background: The idea of studying bone marrow outside its native environment is attractive and ideal. Due to the many functions of extracellular matrix (ECM), currently there is an interest in creating an environment that mimics the ECM present in the tissue, similar to the microenvironment in vivo. Molds replacing the ECM (scaffolds) have a porous structure and may assist the tissue regeneration by forming a suitable environment for adhesion, migration, proliferation and cellular differentiation. The appropriate ECM is a key factor as ECM proteins are site-specific and provide protein 'footprints' of previous resident cells. Because ECM proteins are among the most conserved proteins, the removal of xenogenic/allogenic cellular contents via decellularization could theoretically produce an essentially minimally immunogenic scaffold with a native intact structure for new tissue regeneration. Thus, the search for a scaffold that could be used to assess the behavior of cells and their interactions with the ECM in vitro/in vivo, and has different niches in its composition is highly desirable. Aims: In recent years, a large number of molecular and cytogenetic abnormalities have been identified in AML, MDS and multiple myeloma, many of these defects can serve as markers for diagnosis/prognosis or as therapeutic targets. However, there are still many unknown molecular factors involved in genetic abnormalities or signaling pathways that contribute to the pathogenesis of the disease. Another very important aspect of these diseases is that they all are related to the mutual interaction of neoplastic cells and the microenvironment of bone marrow. In the absence of an ideal model or even the difficulty in reproduce a native environment, we proposed the characterization of a natural scaffold, from bovine bone marrow, which can be used as a study model, previously patented by our laboratory. Materials and Methods: Bone marrow was decellularized by one or more incubations in an enzymatic digestion solution and polar solvent extractions, comprising an extracellular matrix with well-preserved 3D structure. Scaffolds were analyzed after the decelularization process for potential changes in structure (TEM, SEM, Histological staining, and immunohistochemistry for collagen III, IV, fibronectin) and mechanical properties. To verify if the scaffold would hold and support cell survival and extracellular matrix production, an in vitro study was performed using CD34+ (non-stromal) and HS-5 (stromal) cells. Cell-seeded decellularized scaffolds were cultured for 7-14 days and analyzed for Histological staining. Results: Histology sections (H&E staining), TEM and SEM demonstrated the structure and ultrastructure of the processed matrix and confirmed both cellular extraction and preservation of the macroscopic 3-D architecture of the collagen fibers, blood vessels, and preservation of an organized matrix. Also, the decellularized scaffold was quite comparable to the native tissue in terms of its mechanical properties. Immunohistochemistry of the scaffold showed that the main components of the ECM were preserved. The in vitro experiments of both stromal cells (HS-5) and non-stromal cells (CD34+) demonstrated that they were able to adhere and in the HS-5 case also produce ECM during 7-14 days of culture. In both cases, an increase in cell number was observed and CD34+ overtime formed cluster and with 14 days of culture the cluster formation increased in size. Conclusions: The results demonstrated that the decellularization process was efficient in keeping a 3-D structure and mechanical properties with a well-organized-preserved ECM. In vitro experiments showed that both CD34+ and HS-5 were able to proliferate and adhere in specific sites of the scaffold, suggesting that they were able to recognize their native environment. HS-5 produced ECM indicating that the scaffold worked as an optimal microenvironment. In conclusion, the scaffold could be used as a model, which has the potential to mimic the native microenvironment to enable research/studies of factors that are involved in self-renewal and maintenance of neoplastic cells in bone marrow. Also, this model could be very useful for pharmacological testing of bone marrow in vitro. Disclosures No relevant conflicts of interest to declare.


2013 ◽  
Vol 2013 ◽  
pp. 1-35 ◽  
Author(s):  
Patricia Zuk

In 2001, researchers at the University of California, Los Angeles, described the isolation of a new population of adult stem cells from liposuctioned adipose tissue. These stem cells, now known as adipose-derived stem cells or ADSCs, have gone on to become one of the most popular adult stem cells populations in the fields of stem cell research and regenerative medicine. As of today, thousands of research and clinical articles have been published using ASCs, describing their possible pluripotency in vitro, their uses in regenerative animal models, and their application to the clinic. This paper outlines the progress made in the ASC field since their initial description in 2001, describing their mesodermal, ectodermal, and endodermal potentials both in vitro and in vivo, their use in mediating inflammation and vascularization during tissue regeneration, and their potential for reprogramming into induced pluripotent cells.


2013 ◽  
Vol 51 (3) ◽  
pp. T75-T85 ◽  
Author(s):  
Stefania Carobbio ◽  
Barry Rosen ◽  
Antonio Vidal-Puig

Confirmation of the presence of functional brown adipose tissue (BAT) in humans has renewed interest in investigating the potential therapeutic use of this tissue. The finding that its activity positively correlates with decreased BMI, decreased fat content, and augmented energy expenditure suggests that increasing BAT mass/activity or browning of white adipose tissue (WAT) could be a strategy to prevent or treat obesity and its associated morbidities. The challenge now is to find a safe and efficient way to develop this idea. Whereas BAT has being widely studied in murine models bothin vivoandin vitro, there is an urgent need for human cellular models to investigate BAT physiology and functionality from a molecular point of view. In this review, we focus on the latest insights surrounding BAT development and activation in rodents and humans. Then, we discuss how the availability of murine models has been essential to identify BAT progenitors and trace their lineage. Finally, we address how this information can be exploited to develop human cellular models for BAT differentiation/activation. In this context, human embryonic stem and induced pluripotent stem cells-based cellular models represent a resource of great potential value, as they can provide a virtually inexhaustible supply of starting material for functional genetic studies, -omics based analysis and validation of therapeutic approaches. Moreover, these cells can be readily genetically engineered, opening the possibility of generating patient-specific cellular models, allowing the investigation of the influence of different genetic backgrounds on BAT differentiation in pathological or in physiological states.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Pengyu Hong ◽  
Xiaoyang Xu ◽  
Xin Hu ◽  
Hao Yang ◽  
Yue Wu ◽  
...  

Abstract Objective To explore the adipogenic effects of the small extracellular vesicles derived from the lipoma tissues (sEV-LT), and to find a new cell-free therapeutic approach for adipose tissue regeneration. Methods Adipose tissue-derived stem cells (ADSCs) and small extracellular vesicles derived from the adipose tissues (sEV-AT) were isolated from human adipose tissue, while sEV-LT were isolated from human lipomatous tissue. ADSCs were characterized by using flow cytometric analysis and adipogenic and osteogenic differentiation assays. sEV was identified by electron microscopy, nanoparticle tracking, and western blotting. ADSCs were treated with sEV-LT and sEV-AT, respectively. Fluorescence confocal microscopy was used to investigate whether sEV-LT and sEV-AT could be taken by ADSCs. The proliferation and migration abilities and adipogenic differentiation assay of ADSCs were evaluated by CCK-8 assays, scratch test, and oil red O staining test, and the expression levels of adipogenic-related genes C/EBP-δ, PPARγ2, and Adiponectin in ADSCs were assessed by real-time quantitative PCR (RT-PCR). The sEV-LT and sEV-AT transplantation tubes were implanted subcutaneously in SD rats, and the neotissues were qualitatively and histologically evaluated at 2, 4, 8, and 12 weeks after transplantation. Hematoxylin and eosin (H&E) staining was subsequently used to observe and compare the adipogenesis and angiogenesis in neotissues, while immunohistochemistry was used to examine the expression and the distribution of C/EBP-α, PPARγ, Adiponectin, and CD31 at the 4th week. Results The in vitro experiments showed that both sEV-LT and sEV-AT could be taken up by ADSCs via endocytosis. The scratch experiment and CCK-8 experiment showed that the migration area and proliferation number of ADSCs in sEV-LT group and sEV-AT group were significantly higher than those in the non-sEV group (p < 0.05). Compared with sEV-AT group, sEV-LT group had larger migration area and proliferation number of ADSCs (p < 0.05). Oil red O staining and RT-PCR experiments showed that, compared with the non-sEVs group, the lipid droplets and the mRNA expression levels of adipogenesis-related genes PPARγ2 and Adiponectin of ADSCs in sEV-LT group and sEV-AT group were significantly upregulated (p < 0.05); however, there was no statistical significance in the expression level of C/EBP-δ (p > 0.05). In addition, no significant difference in the amount of lipid droplets and adipogenesis-related genes between the sEV-LT groups and sEV-AT was seen (p > 0.05). At 2, 4, 8, and 12 weeks, the adipocyte area and the number of capillaries in neotissues in the sEV-LT groups and sEV-AT groups were significantly increased compared with the Matrigel group (p < 0.05); however, there was no dramatic difference between sEV-LT groups and sEV-AT groups (p > 0.05). At the 4th week, neotissues in the sEV-LT groups and sEV-AT groups all showed upregulated expression of C/EBP-α, PPARγ, Adiponectin, and CD31 protein, while neotissues in the Matrigel group only showed positive expression of CD31 protein. Conclusions This study demonstrated that sEV-LT exerted promotion effects on adipose tissue regeneration by accelerating the proliferation, migration, and adipogenic differentiation of ADSCs in vitro and recruiting adipocytes and promoting angiogenesis in vivo. The sEV-LT could serve as an alternative cell-free therapeutic strategy for generating adipose tissue, thus providing a promising application prospect in tissue engineering.


2021 ◽  
Vol 0 (0) ◽  
Author(s):  
Matthias Schnabelrauch ◽  
Jürgen Schiller ◽  
Stephanie Möller ◽  
Dieter Scharnweber ◽  
Vera Hintze

Abstract Tissue regeneration is regulated by the cellular microenvironment, e.g. the extracellular matrix. Here, sulfated glycosaminoglycans (GAG), are of vital importance interacting with mediator proteins and influencing their biological activity. Hence, they are promising candidates for controlling tissue regeneration. This review addresses recent achievements regarding chemically modified GAG as well as collagen/GAG-based coatings and hydrogels including (i) chemical functionalization strategies for native GAG, (ii) GAG-based biomaterial strategies for controlling cellular responses, (iii) (bio)chemical methods for characterization and iv) protein interaction profiles and attained tissue regeneration in vitro and in vivo. The potential of GAG for bioinspired, functional biomaterials is highlighted.


2021 ◽  
Author(s):  
Keel Yong Lee ◽  
Huong Nguyen ◽  
Agustina Setiawati ◽  
So-Jung Nam ◽  
Minyoung Kim ◽  
...  

Abstract The unfolded states of fibronectin (FN) subsequently induce the formation of the extracellular matrix (ECM) fibrillar network, which is necessary to generate new substitutive tissues. Here, we demonstrate that negatively charged small unilamellar vesicles (SUVs) qualify as candidates for FN delivery due to their remarkable effects on the autonomous binding and unfolding of FN, which leads to increased tissue regeneration. In vitro experiments revealed that the FN-SUV complex remarkably increased the attachment, differentiation, and migration of fibroblasts. The potential utilization of this complex in vivo to treat inflammatory colon diseases is also described based on results obtained for ameliorated conditions in rats with ulcerative colitis (UC) that had been treated with the FN-SUV complex. Our findings provide a new ECM-delivery platform for ECM-based therapeutic applications and suggest that properly designed SUVs could be an unprecedented FN-delivery system that is highly effective in treating UC and other diseases.


2022 ◽  
Vol 119 (2) ◽  
pp. e2116865118
Author(s):  
Shiv Shah ◽  
Caldon Jayson Esdaille ◽  
Maumita Bhattacharjee ◽  
Ho-Man Kan ◽  
Cato T. Laurencin

Stem cells are of great interest in tissue regeneration due to their ability to modulate the local microenvironment by secreting bioactive factors (collectively, secretome). However, secretome delivery through conditioned media still requires time-consuming cell isolation and maintenance and also may contain factors antagonistic to targeted tissue regeneration. We have therefore engineered a synthetic artificial stem cell (SASC) system which mimics the paracrine effect of the stem cell secretome and provides tailorability of the composition for targeted tissue regeneration. We report the first of many applications of the SASC system we have formulated to treat osteoarthritis (OA). Choosing growth factors important to chondrogenesis and encapsulating respective recombinant proteins in poly (lactic-coglycolic acid) 85:15 (PLGA) we fabricated the SASC system. We compared the antiinflammatory and chondroprotective effects of SASC to that of adipose-derived stem cells (ADSCs) using in vitro interleukin 1B-induced and in vivo collagenase-induced osteoarthritis rodent models. We have designed SASC as an injectable therapy with controlled release of the formulated secretome. In vitro, SASC showed significant antiinflammatory and chondroprotective effects as seen by the up-regulation of SOX9 and reduction of nitric oxide, ADAMTS5, and PRG4 genes compared to ADSCs. In vivo, treatment with SASC and ADSCs significantly attenuated cartilage degeneration and improved the biomechanical properties of the articular cartilage in comparison to OA control. This SASC system demonstrates the feasibility of developing a completely synthetic, tailorable stem cell secretome which reinforces the possibility of developing a new therapeutic strategy that provides better control over targeted tissue engineering applications.


Sign in / Sign up

Export Citation Format

Share Document