Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Tissue Eng Part A ; 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38874518

ABSTRACT

Cell aggregates are widely used to study heterotypic cellular interactions during the development of vascularization in vitro. In this study, we examined heterotypic cellular spheroids made of adipose-derived stem cells and CD34+/CD31- endothelial progenitor cells induced by the transfection of miR-148b mimic for de novo induction of osteogenic differentiation and miR-210 mimic for de novo induction of endotheliogenesis, respectively. The effect of the microRNA (miRs) mimic treatment group and induction time on codifferentiation was assessed in spheroids formed of transfected cells over the course of a 4-week culture. Based on gene and protein markers of osteogenic and endotheliogenic differentiation, as well as mineralization assays, our results showed that miRs directed cell differentiation and that progenitor maturity influenced the development of heterotypic cellular regions in aggregates. Overall, the success of coculture to create a prevascularized bone model is dependent on a number of factors, particularly the induction time of differentiation before combining the multiple cell types in aggregates. The approach that has been proposed could be valuable in creating vascularized bone tissue by employing spheroids as the building blocks of more complex issues through the use of cutting-edge methods such as 3D bioprinting.

2.
Minerva Surg ; 79(2): 166-182, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38088753

ABSTRACT

Tissue loss, irrespective of etiology, often requires extensive reconstruction. In many instances, the need exceeds what current treatments and technologies modern medicine can offer. Tissue engineering has made immense strides within the past few decades due to advances in biologics, biomaterials, and manufacturing. The convergence of these three domains has created limitless potential for future surgical care. Unfortunately, there still exists a disconnect on how to best implant these 'replacement parts' and care for the patient. It is therefore vital to develop paradigms for the integration of advanced surgical and tissue engineering technologies. This paper explores the convergence between tissue engineering and reconstructive surgery. We will describe the clinical problem of tissue loss, discuss currently available solutions, address limitations, and propose processes for integrating surgery and tissue engineering, thereby ushering in the era of regenerative surgery.


Subject(s)
Biocompatible Materials , Tissue Engineering , Humans , Biocompatible Materials/therapeutic use , Tissue Scaffolds
3.
Macromol Biosci ; 24(3): e2300475, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37955619

ABSTRACT

Hydrogels loaded with biologics hold great potential for various biomedical applications such as regenerative medicine. However, biologics may lose bioactivity during hydrogel preparation, shipping, and storage. While many injectable hydrogels do not have this issue, they face a dilemma between fast gelation causing the difficulty of injection and slow gelation causing the escape of solutions from an injection site. The purpose of this study is to develop an affinity hydrogel by integrating a pre-formed elastic macroporous matrix and an injectable hydrogel. The data shows that the macroporous hydrogel matrix can hold a large volume of solutions for the formation of in situ injectable hydrogels loaded with growth factors or living cells. The cells can proliferate in the composite hydrogels. The growth factors can be stably sequestered and sustainably released due to the presence of aptamers. When both living cells and growth factors are loaded together into the hydrogels, cells can proliferate under culture conditions with a reduced serum level. Therefore, a macroporous and elastic matrix-supported formation of aptamer-functionalized injectable hydrogels is a promising method for developing the carriers of biologics.


Subject(s)
Biological Products , Hydrogels , Hydrogels/pharmacology , Intercellular Signaling Peptides and Proteins , Regenerative Medicine , Extracellular Matrix
4.
Microcirculation ; 31(1): e12835, 2024 01.
Article in English | MEDLINE | ID: mdl-37947797

ABSTRACT

OBJECTIVE: The success of engineered tissues continues to be limited by time to vascularization and perfusion. Recently, we described a simple microsurgical approach, termed micropuncture (MP), which could be used to rapidly vascularize an adjacently placed scaffold from the recipient macrovasculature. Here we studied the long-term persistence of the MP-induced microvasculature. METHODS: Segmental 60 µm diameter MPs were created in the recipient rat femoral artery and vein followed by coverage with a simple Type 1 collagen scaffold. The recipient vasculature and scaffold were then wrapped en bloc with a silicone sheet to isolate intrinsic vascularization. Scaffolds were harvested at 28 days post-implantation for detailed analysis, including using a novel artificial intelligence (AI) approach. RESULTS: MP scaffolds demonstrated a sustained increase of vascular density compared to internal non-MP control scaffolds (p < 0.05) secondary to increases in both vessel diameters (p < 0.05) and branch counts (p < 0.05). MP scaffolds also demonstrated statistically significant increases in red blood cell (RBC) perfused lumens. CONCLUSIONS: This study further highlights that the intrinsic MP-induced vasculature continues to persist long-term. Its combination of rapid and stable angiogenesis represents a novel surgical platform for engineered scaffold and graft perfusion.


Subject(s)
Artificial Intelligence , Tissue Scaffolds , Animals , Rats , Punctures , Silicones , Tissue Engineering , Angiogenesis
5.
Small ; 20(8): e2307928, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37824280

ABSTRACT

Bulk hydrogel scaffolds are common in reconstructive surgery. They allow for the staged repair of soft tissue loss by providing a base for revascularization. Unfortunately, they are limited by both slow and random vascularization, which may manifest as treatment failure or suboptimal repair. Rapidly inducing patterned vascularization within biomaterials has profound translational implications for current clinical treatment paradigms and the scaleup of regenerative engineering platforms. To address this long-standing challenge, a novel microsurgical approach and granular hydrogel scaffold (GHS) technology are co-developed to hasten and pattern microvascular network formation. In surgical micropuncture (MP), targeted recipient blood vessels are perforated using a microneedle to accelerate cell extravasation and angiogenic outgrowth. By combining MP with an adjacent GHS with precisely tailored void space architecture, microvascular pattern formation as assessed by density, diameter, length, and intercapillary distance is rapidly guided. This work opens new translational opportunities for microvascular engineering, advancing reconstructive surgery, and regenerative medicine.


Subject(s)
Tissue Engineering , Tissue Scaffolds , Humans , Hydrogels/pharmacology , Neovascularization, Pathologic , Punctures , Neovascularization, Physiologic
6.
Bioact Mater ; 33: 114-128, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38024230

ABSTRACT

Craniomaxillofacial (CMF) reconstruction is a challenging clinical dilemma. It often necessitates skin replacement in the form of autologous graft or flap surgery, which differ from one another based on hypodermal/dermal content. Unfortunately, both approaches are plagued by scarring, poor cosmesis, inadequate restoration of native anatomy and hair, alopecia, donor site morbidity, and potential for failure. Therefore, new reconstructive approaches are warranted, and tissue engineered skin represents an exciting alternative. In this study, we demonstrated the reconstruction of CMF full-thickness skin defects using intraoperative bioprinting (IOB), which enabled the repair of defects via direct bioprinting of multiple layers of skin on immunodeficient rats in a surgical setting. Using a newly formulated patient-sourced allogenic bioink consisting of both human adipose-derived extracellular matrix (adECM) and stem cells (ADSCs), skin loss was reconstructed by precise deposition of the hypodermal and dermal components under three different sets of animal studies. adECM, even at a very low concentration such as 2 % or less, has shown to be bioprintable via droplet-based bioprinting and exhibited de novo adipogenic capabilities both in vitro and in vivo. Our findings demonstrate that the combinatorial delivery of adECM and ADSCs facilitated the reconstruction of three full-thickness skin defects, accomplishing near-complete wound closure within two weeks. More importantly, both hypodermal adipogenesis and downgrowth of hair follicle-like structures were achieved in this two-week time frame. Our approach illustrates the translational potential of using human-derived materials and IOB technologies for full-thickness skin loss.

7.
bioRxiv ; 2023 Oct 05.
Article in English | MEDLINE | ID: mdl-37873077

ABSTRACT

Craniomaxillofacial (CMF) reconstruction is a challenging clinical dilemma. It often necessitates skin replacement in the form of autologous graft or flap surgery, which differ from one another based on hypodermal/dermal content. Unfortunately, both approaches are plagued by scarring, poor cosmesis, inadequate restoration of native anatomy and hair, alopecia, donor site morbidity, and potential for failure. Therefore, new reconstructive approaches are warranted, and tissue engineered skin represents an exciting alternative. In this study, we demonstrated the reconstruction of CMF full-thickness skin defects using intraoperative bioprinting (IOB), which enabled the repair of defects via direct bioprinting of multiple layers of skin on immunodeficient rats in a surgical setting. Using a newly formulated patient-sourced allogenic bioink consisting of both human adipose-derived extracellular matrix (adECM) and stem cells (ADSCs), skin loss was reconstructed by precise deposition of the hypodermal and dermal components under three different sets of animal studies. adECM, even at a very low concentration such as 2% or less, has shown to be bioprintable via droplet-based bioprinting and exhibited de novo adipogenic capabilities both in vitro and in vivo . Our findings demonstrate that the combinatorial delivery of adECM and ADSCs facilitated the reconstruction of three full-thickness skin defects, accomplishing near-complete wound closure within two weeks. More importantly, both hypodermal adipogenesis and downgrowth of hair follicle-like structures were achieved in this two-week time frame. Our approach illustrates the translational potential of using human-derived materials and IOB technologies for full-thickness skin loss.

8.
Surg Open Sci ; 11: 73-76, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36578695

ABSTRACT

Background: Innovation is broadly defined as the act of introducing a new product, idea, or process. The field of surgery is built upon innovation, revolutionizing technology, science, and tools to improve patient care. While most innovative solutions are aimed at problems with a significant patient population, the process can also be used on orphan pathologies without obvious solutions. We present a case of tracheal agenesis, a rare congenital anomaly with an overwhelming mortality and few good treatment options, that benefited from the innovation process and achieved survival with no ventilator dependence at three years of age. Methods: Utilizing the framework of the innovation process akin to the Stanford Biodesign Program, 1) the parameters of the clinical problem were identified, 2) previous solutions and existing technologies were analyzed, newly invented solutions were brainstormed, and value analysis of the possible solutions were carried out using crowd wisdom, and 3) the selected solution was prototyped and tested using 3D modeling, iterative testing on 3D prints of actual-sized patient parts, and eventual implementation in the patient after regulatory clearance. Results: A 3D-printed external bioresorbable splint was chosen as the solution. Our patient underwent airway reconstruction with "trachealization of the esophagus": esophageotracheal fistula resection, esophagotracheoplasty, and placement of a 3D-printed polycaprolactone (PCL) stent for external esophageal airway support at five months of age. Conclusions: The innovation process provided our team with the guidance and imperative steps necessary to develop an innovative device for the successful management of an infant survivor with Floyd Type I tracheal agenesis. Article summary: We present a case of tracheal agenesis, a rare congenital anomaly with an overwhelming mortality and few good treatment options, that benefited from the innovation process and achieved survival with no ventilator dependence at three years of age.The importance of this report is to reveal how the innovation process, which is typically used for problems with significant patient population, can also be used on orphan pathologies without obvious solutions.

9.
Support Care Cancer ; 30(12): 10301-10310, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36355217

ABSTRACT

PURPOSE: Ability to return to work (RTW) is an important aspect of breast cancer that is limited for many survivors. With 90% survivorship in the USA, it is imperative that focus shifts toward the improvement of physical arm function to improve survivors' ability to RTW. This narrative review discusses the role of physical arm function and demographic disparities in breast cancer survivor RTW. METHODS: Literature on physical function, arm function, and demographic disparities following breast cancer treatment and their implications for RTW is discussed. RESULTS: The ability to RTW is a key component of recovery for breast cancer survivors, but challenges and inequalities persist. Treatment effects can induce and prolong functional disability, affecting survivors' ability to RTW. These effects may be compounded for survivors whose occupation requires physical arm function. The RTW landscape, including the occupations survivors have, the physical function required for job tasks, and availability of workplace accommodations, is also unclear. Additional demographic disparities (e.g., income, live in rural area) exist, but the extent to which these factors influence RTW is not well understood. More work is needed to understand the compounded impact of treatment effects, demographic disparities, and occupational factors on RTW. Multidisciplinary rehabilitation that includes occupational counseling and exercise is a promising approach, but widespread adoption in the US healthcare model presents an ongoing challenge. Areas for further research are highlighted. CONCLUSION: There is an incomplete understanding of the effects of treatment on physical arm function and the role of demographic disparities on breast cancer survivor RTW.


Subject(s)
Breast Neoplasms , Cancer Survivors , Humans , Female , Cancer Survivors/psychology , Return to Work/psychology , Breast Neoplasms/psychology , Arm , Survivors/psychology , Demography
10.
Cells ; 11(21)2022 10 25.
Article in English | MEDLINE | ID: mdl-36359762

ABSTRACT

Extracellular vesicles (EVs) are small lipid bilayer-delimited particles that are naturally released from cells into body fluids, and therefore can travel and convey regulatory functions in the distal parts of the body. EVs can transmit paracrine signaling by carrying over cytokines, chemokines, growth factors, interleukins (ILs), transcription factors, and nucleic acids such as DNA, mRNAs, microRNAs, piRNAs, lncRNAs, sn/snoRNAs, mtRNAs and circRNAs; these EVs travel to predecided destinations to perform their functions. While mesenchymal stem cells (MSCs) have been shown to improve healing and facilitate treatments of various diseases, the allogenic use of these cells is often accompanied by serious adverse effects after transplantation. MSC-produced EVs are less immunogenic and can serve as an alternative to cellular therapies by transmitting signaling or delivering biomaterials to diseased areas of the body. This review article is focused on understanding the properties of EVs derived from different types of MSCs and MSC-EV-based therapeutic options. The potential of modern technologies such as 3D bioprinting to advance EV-based therapies is also discussed.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , MicroRNAs , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , Cell- and Tissue-Based Therapy , MicroRNAs/genetics , MicroRNAs/metabolism , Bioengineering
11.
Ann Plast Surg ; 88(3 Suppl 3): S288-S292, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35513333

ABSTRACT

INTRODUCTION: Ongoing recognition of breast implant-associated anaplastic large cell lymphoma (BIA-ALCL) and its link with textured devices has brought a paradigm shift in prosthetic-based breast reconstruction. Many institutions no longer offer textured expansion devices for staged reconstruction. However, there is a paucity of data regarding the efficacy of smooth tissue expanders (TE). We hypothesized that the time to final reconstruction and complication profile between smooth and textured TEs would be similar in breast reconstruction patients. METHODS: A retrospective chart review was performed of all patients who underwent TE breast reconstruction during a 6-year period at the Penn State Hershey Medical Center. Rates of complications treated nonoperatively and those requiring reoperation were assessed. Mechanical complications, including expander malposition and rupture, were evaluated. Time to final breast reconstruction was quantified. Mixed-effects logistic regression and linear regression models, as appropriate, were used to compare textured to smooth TEs. Patient characteristics and anatomic plane placement were adjusted for in all analyses of outcomes. RESULTS: Data were collected on 389 patients, encompassing 140 smooth and 604 textured TEs. Textured devices had an increased incidence of complications treated nonsurgically (16.7% vs 10.7%; P = 0.14). However, smooth TEs had an increased incidence of reoperation (12.1% vs 7.6%; P = 0.06). Most noteworthy was that although smooth TEs had a 40-fold increase in malposition (13.6% vs 0.3%; P < 0.001), no reoperation for this complication was warranted. Further, the time to final reconstruction was comparable between the 2 devices (textured expanders: 221 days and smooth expanders: 234 days; P = 0.15). CONCLUSIONS: Staged, implant-based reconstruction is the most common surgical approach to recreate the breast mound following mastectomy. Textured TEs were the cornerstone to this approach. Unfortunately, the association between textured devices and BIA-ALCL now mandates an alternative. We postulated that smooth expanders would compare favorably for breast reconstruction. Although our study suggests that smooth TEs suffer more malposition, this has a negligible impact on the reconstructive timeline. Thus, smooth TEs may prove beneficial when considering the risk of BIA-ALCL associated with textured devices.


Subject(s)
Breast Implantation , Breast Implants , Breast Neoplasms , Lymphoma, Large-Cell, Anaplastic , Mammaplasty , Breast Implantation/adverse effects , Breast Implants/adverse effects , Breast Neoplasms/complications , Breast Neoplasms/surgery , Female , Humans , Lymphoma, Large-Cell, Anaplastic/epidemiology , Lymphoma, Large-Cell, Anaplastic/etiology , Lymphoma, Large-Cell, Anaplastic/surgery , Mammaplasty/adverse effects , Mastectomy/adverse effects , Postoperative Complications/epidemiology , Postoperative Complications/etiology , Postoperative Complications/surgery , Retrospective Studies , Tissue Expansion Devices/adverse effects
12.
J Biomed Mater Res A ; 110(1): 156-163, 2022 01.
Article in English | MEDLINE | ID: mdl-34263999

ABSTRACT

Decellularized extracellular matrix (ECM) from tissues is a promising biomaterial that can provide a complex 3D microenvironment capable of modulating cell response and tissue regeneration. In this study, we have integrated the decellularized thiolated adipose-derived ECM, at different concentrations, with polyethylene glycol (PEG) using Michael addition between thiol and acrylate moieties. The potential for this material to support adipogenic differentiation of human adipose-derived stem cells was evaluated by encapsulating cells in hydrogels with increasing concentrations of chemically modified ECM (mECM). Our results demonstrated a positive correlation between the ECM content in the hydrogels and cell proliferation, adipogenic marker expression, and lipid formation and accumulation. Furthermore, we have shown host cell infiltration and enhanced adipogenesis in vivo after implantation. These findings support the graft as a potential alternative for adipose tissue regeneration.


Subject(s)
Extracellular Matrix , Tissue Scaffolds , Adipogenesis , Adipose Tissue , Biocompatible Materials/chemistry , Extracellular Matrix/chemistry , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Tissue Engineering/methods , Tissue Scaffolds/chemistry
13.
Front Surg ; 8: 731031, 2021.
Article in English | MEDLINE | ID: mdl-34805257

ABSTRACT

Many pathologies, congenital defects, and traumatic injuries are untreatable by conventional pharmacologic or surgical interventions. Regenerative engineering represents an ever-growing interdisciplinary field aimed at creating biological replacements for injured tissues and dysfunctional organs. The need for bioengineered replacement parts is ubiquitous among all surgical disciplines. However, to date, clinical translation has been limited to thin, small, and/or acellular structures. Development of thicker tissues continues to be limited by vascularization and other impediments. Nevertheless, currently available materials, methods, and technologies serve as robust platforms for more complex tissue fabrication in the future. This review article highlights the current methodologies, clinical achievements, tenacious barriers, and future perspectives of regenerative engineering.

14.
Stem Cells Dev ; 30(23): 1153-1170, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34514867

ABSTRACT

Diabetes is a pandemic manifested through glucose dysregulation mediated by inadequate insulin secretion by beta cells. A beta cell replacement strategy would transform the treatment paradigm from pharmacologic glucose modulation to a genuine cure. Stem cells have emerged as a potential source for beta cell (ß-cell) engineering. The detailed generation of functional ß-cells from both embryonic and induced pluripotent stem cells has recently been described. Adult stem cells, including adipose derived, may also offer a therapeutic approach, but remain ill defined. In our study, we performed an in-depth assessment of insulin-producing beta cells generated from human adipose, irrespective of donor patient age, gender, and health status. Cellular transformation was confirmed using flow cytometry and single-cell imaging. Insulin secretion was observed with glucose stimulation and abrogated following palmitate exposure, a common free fatty acid implicated in human beta cell dysfunction. We used next-generation sequencing to explore gene expression changes before and after differentiation of patient-matched samples, which revealed more than 5,000 genes enriched. Adipose-derived beta cells displayed comparable gene expression to native ß-cells. Pathway analysis demonstrated relevance to stem cell differentiation and pancreatic developmental processes, which are vital to cellular function, structural development, and regulation. We conclude that the functions associated with adipose derived beta cells are mediated through relevant changes in the transcriptome, which resemble those seen in native ß-cell morphogenesis and maturation. Therefore, they may represent a viable option for the clinical translation of stem cell-based therapies in diabetes.


Subject(s)
Induced Pluripotent Stem Cells , Insulin-Secreting Cells , Cell Differentiation/genetics , Genomics , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism
15.
Adv Funct Mater ; 31(29)2021 Jul 16.
Article in English | MEDLINE | ID: mdl-34421475

ABSTRACT

Reconstruction of complex craniomaxillofacial (CMF) defects is challenging due to the highly organized layering of multiple tissue types. Such compartmentalization necessitates the precise and effective use of cells and other biologics to recapitulate the native tissue anatomy. In this study, intra-operative bioprinting (IOB) of different CMF tissues, including bone, skin, and composite (hard/soft) tissues, is demonstrated directly on rats in a surgical setting. A novel extrudable osteogenic hard tissue ink is introduced, which induced substantial bone regeneration, with ≈80% bone coverage area of calvarial defects in 6 weeks. Using droplet-based bioprinting, the soft tissue ink accelerated the reconstruction of full-thickness skin defects and facilitated up to 60% wound closure in 6 days. Most importantly, the use of a hybrid IOB approach is unveiled to reconstitute hard/soft composite tissues in a stratified arrangement with controlled spatial bioink deposition conforming the shape of a new composite defect model, which resulted in ≈80% skin wound closure in 10 days and 50% bone coverage area at Week 6. The presented approach will be absolutely unique in the clinical realm of CMF defects and will have a significant impact on translating bioprinting technologies into the clinic in the future.

16.
J Surg Res ; 261: 173-178, 2021 05.
Article in English | MEDLINE | ID: mdl-33444946

ABSTRACT

BACKGROUND: Nasoalveolar molding is a nonsurgical modality for the treatment of cleft lip and palate that uses an intraoral splint to align the palatal shelves. Repeated impressions are needed for splint modification, each carrying risk of airway obstruction. Computer-aided design and manufacturing (CAD/CAM) has the ability to simplify the process. As a precursor to CAD/CAM splint fabrication, a proof-of-concept study was conducted to compare three-dimensional splints printed from alginate impressions versus digital scans. We hypothesized that intraoral digital scanning would compare favorably to alginate impressions for palate registration and subsequent splint manufacture, with decreased production times. METHODS: Alginate and digital impressions were taken from 25 healthy teenage volunteers. Digital impressions were performed with a commercially available intraoral scanner. Plaster casts made from alginate impressions were converted to Standard Triangle Language files. Patient-specific matched scans were evaluated for total surface area with the concordance correlation coefficient. Acrylic palatal splints were three-dimensionally printed from inverse digital molds. Subjective appliance fit was assessed using a five-point scale. RESULTS: A total of 23 participants were included. Most subjects preferred digital impression acquisition. Impression methods showed moderate agreement (concordance correlation coefficient 0.93). Subjects rated splints from digital impressions as having a more precise fit (4.4 versus 3.9). The digital approach decreased impression phase time by over 10-fold and overall production time by 28%. CONCLUSIONS: CAD/CAM has evolved extensively over the past two decades and is now commonplace in medicine. However, its utility in cleft patients has not been fully realized. This pilot study demonstrated that CAD/CAM technologies may prove useful in patients requiring intraoral splints.


Subject(s)
Cleft Palate/therapy , Computer-Aided Design , Diagnosis, Oral/methods , Nasoalveolar Molding/instrumentation , Palatal Obturators , Adolescent , Alginates , Healthy Volunteers , Humans , Pilot Projects , Splints , Young Adult
17.
Microvasc Res ; 134: 104121, 2021 03.
Article in English | MEDLINE | ID: mdl-33309646

ABSTRACT

The success of engineered tissues continues to be limited by time to vascularization and perfusion. Here, we studied the effects of precision injury to a recipient macrovasculature in promoting neovessel formation in an adjacently placed scaffold. Segmental 60 µm diameter micropunctures (MP) were created in the recipient rat femoral artery and vein followed by coverage with a simple collagen scaffold. Scaffolds were harvested at 24, 48, 72, and 96 h post-implantation for detailed analysis. Those placed on top of an MP segment showed an earlier and more robust cellular infiltration, including both endothelial cells (CD31) and macrophages (F4/80), compared to internal non-micropunctured control limbs (p < 0.05). At the 96-hour timepoint, MP scaffolds demonstrated an increase in physiologic perfusion (p < 0.003) and a 2.5-fold increase in capillary network formation (p < 0.001). These were attributed to an overall upsurge in small vessel quantity. Furthermore, MP positioned scaffolds demonstrated significant increases in many modulators of angiogenesis, including VEGFR2 and Tie-2 despite a decrease in HIF-1α at all timepoints. This study highlights a novel microsurgical approach that can be used to rapidly vascularize or inosculate contiguously placed scaffolds and grafts. Thereby, offering an easily translatable route towards the creation of thicker and more clinically relevant engineered tissues.


Subject(s)
Femoral Artery , Femoral Vein , Hindlimb/blood supply , Neovascularization, Physiologic , Tissue Engineering , Tissue Scaffolds , Animals , Collagen/metabolism , Femoral Artery/metabolism , Femoral Vein/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Punctures , Rats, Sprague-Dawley , Receptor, TIE-2/metabolism , Signal Transduction , Time Factors , Vascular Endothelial Growth Factor Receptor-2/metabolism
18.
Regen Eng Transl Med ; 6(1): 101-110, 2020 Mar.
Article in English | MEDLINE | ID: mdl-33344757

ABSTRACT

In this study, CD34+/CD31- progenitor cells were isolated from the stromal vascular fraction (SVF) of adipose tissue using magnetic activated cell sorting. The endothelial differentiation capability of these cells in vitro was evaluated by culturing them in vascular endothelial growth factor (VEGF) induced medium for 14 days. Viability, proliferation, differentiation and tube formation of these cells were evaluated. Cell viability study revealed that both undifferentiated and endothelial differentiated cells remained healthy for 14 days. However, the proliferation rate was higher in undifferentiated cells compared to endothelial differentiated ones. Upregulation of endothelial characteristic genes (Von Willebrand Factor (vWF) and VE Cadherin) was observed in 2D culture. However, PECAM (CD31) was only found to be upregulated after the cells had formed tube-like structures in 3D Matrigel culture. These results indicate that adipose derived CD34+/CD31- cells when cultured in VEGF induced medium, are capable differentiation into endothelial-like lineages. Tube formation of the cells started 3h after seeding the cells on Matrigel and formed more stable and connected network 24 h post seeding in presence of VEGF.

19.
Adv Healthc Mater ; 9(22): e2001657, 2020 11.
Article in English | MEDLINE | ID: mdl-33073548

ABSTRACT

The heterogeneous and anisotropic articular cartilage is generally studied as a layered structure of "zones" with unique composition and architecture, which is difficult to recapitulate using current approaches. A novel hybrid bioprinting strategy is presented here to generate zonally stratified cartilage. Scaffold-free tissue strands (TSs) are made of human adipose-derived stem cells (ADSCs) or predifferentiated ADSCs. Cartilage TSs with predifferentiated ADSCs exhibit improved mechanical properties and upregulated expression of cartilage-specific markers at both transcription and protein levels as compared to TSs with ADSCs being differentiated in the form of strands and TSs of nontransfected ADSCs. Using the novel hybrid approach integrating new aspiration-assisted and extrusion-based bioprinting techniques, the bioprinting of zonally stratified cartilage with vertically aligned TSs at the bottom zone and horizontally aligned TSs at the superficial zone is demonstrated, in which collagen fibers are aligned with designated orientation in each zone imitating the anatomical regions and matrix orientation of native articular cartilage. In addition, mechanical testing study reveals a compression modulus of ≈1.1 MPa, which is similar to that of human articular cartilage. The prominent findings highlight the potential of this novel bioprinting approach for building biologically, mechanically, and histologically relevant cartilage for tissue engineering purposes.


Subject(s)
Bioprinting , Cartilage, Articular , Tissue Scaffolds , Humans , Stem Cells , Tissue Engineering
20.
Trends Biotechnol ; 38(6): 594-605, 2020 06.
Article in English | MEDLINE | ID: mdl-32407688

ABSTRACT

3D bioprinting directly into injured sites in a surgical setting, intraoperative bioprinting (IOB), is an effective process, in which the defect information can be rapidly acquired and then repaired via bioprinting on a live subject. In patients needing tissue resection, debridement, traumatic reconstruction, or fracture repair, the ability to scan and bioprint immediately following surgical preparation of the defect site has great potential to improve the precision and efficiency of these procedures. In this opinion article, we provide the reader with current major limitations of IOB from engineering and clinical points of view, as well as possibilities of future translation of bioprinting technologies from bench to bedside, and expound our perspectives in the context of IOB of composite and vascularized tissues.


Subject(s)
Bioprinting/trends , Printing, Three-Dimensional/trends , Tissue Engineering/trends , Humans , Tissue Scaffolds/trends
SELECTION OF CITATIONS
SEARCH DETAIL
...