Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Lancet Child Adolesc Health ; 6(9): 654-666, 2022 09.
Article in English | MEDLINE | ID: covidwho-20243577

ABSTRACT

Paper 2 of the paediatric regenerative medicine Series focuses on recent advances in postnatal approaches. New gene, cell, and niche-based technologies and their combinations allow structural and functional reconstitution and simulation of complex postnatal cell, tissue, and organ hierarchies. Organoid and tissue engineering advances provide human disease models and novel treatments for both rare paediatric diseases and common diseases affecting all ages, such as COVID-19. Preclinical studies for gastrointestinal disorders are directed towards oesophageal replacement, short bowel syndrome, enteric neuropathy, biliary atresia, and chronic end-stage liver failure. For respiratory diseases, beside the first human tracheal replacement, more complex tissue engineering represents a promising solution to generate transplantable lungs. Genitourinary tissue replacement and expansion usually involve application of biocompatible scaffolds seeded with patient-derived cells. Gene and cell therapy approaches seem appropriate for rare paediatric diseases of the musculoskeletal system such as spinal muscular dystrophy, whereas congenital diseases of complex organs, such as the heart, continue to challenge new frontiers of regenerative medicine.


Subject(s)
COVID-19 , Regenerative Medicine , Child , Humans , Tissue Engineering
2.
Stem Cell Res Ther ; 14(1): 112, 2023 04 27.
Article in English | MEDLINE | ID: covidwho-2323672

ABSTRACT

Cell therapy is an accessible method for curing damaged organs or tissues. Yet, this approach is limited by the delivery efficiency of cell suspension injection. Over recent years, biological scaffolds have emerged as carriers of delivering therapeutic cells to the target sites. Although they can be regarded as revolutionary research output and promote the development of tissue engineering, the defect of biological scaffolds in repairing cell-dense tissues is apparent. Cell sheet engineering (CSE) is a novel technique that supports enzyme-free cell detachment in the shape of a sheet-like structure. Compared with the traditional method of enzymatic digestion, products harvested by this technique retain extracellular matrix (ECM) secreted by cells as well as cell-matrix and intercellular junctions established during in vitro culture. Herein, we discussed the current status and recent progress of CSE in basic research and clinical application by reviewing relevant articles that have been published, hoping to provide a reference for the development of CSE in the field of stem cells and regenerative medicine.


Subject(s)
Regenerative Medicine , Tissue Engineering , Regenerative Medicine/methods , Tissue Engineering/methods , Cell Engineering , Stem Cells , Cell- and Tissue-Based Therapy , Extracellular Matrix , Tissue Scaffolds
3.
Int J Mol Sci ; 24(7)2023 Mar 23.
Article in English | MEDLINE | ID: covidwho-2291253

ABSTRACT

Cultivated meat (CM) technology has the potential to disrupt the food industry-indeed, it is already an inevitable reality. This new technology is an alternative to solve the environmental, health and ethical issues associated with the demand for meat products. The global market longs for biotechnological improvements for the CM production chain. CM, also known as cultured, cell-based, lab-grown, in vitro or clean meat, is obtained through cellular agriculture, which is based on applying tissue engineering principles. In practice, it is first necessary to choose the best cell source and type, and then to furnish the necessary nutrients, growth factors and signalling molecules via cultivation media. This procedure occurs in a controlled environment that provides the surfaces necessary for anchor-dependent cells and offers microcarriers and scaffolds that favour the three-dimensional (3D) organisation of multiple cell types. In this review, we discuss relevant information to CM production, including the cultivation process, cell sources, medium requirements, the main obstacles to CM production (consumer acceptance, scalability, safety and reproducibility), the technological aspects of 3D models (biomaterials, microcarriers and scaffolds) and assembly methods (cell layering, spinning and 3D bioprinting). We also provide an outlook on the global CM market. Our review brings a broad overview of the CM field, providing an update for everyone interested in the topic, which is especially important because CM is a multidisciplinary technology.


Subject(s)
Meat Products , Tissue Engineering , Tissue Engineering/methods , Reproducibility of Results , Meat , Biotechnology , Tissue Scaffolds
4.
Int J Mol Sci ; 24(6)2023 Mar 19.
Article in English | MEDLINE | ID: covidwho-2256018

ABSTRACT

In December 2019, COVID-19 emerged in China, and in January 2020, the World Health Organization declared a state of international emergency. Within this context, there is a significant search for new drugs to fight the disease and a need for in vitro models for preclinical drug tests. This study aims to develop a 3D lung model. For the execution, Wharton's jelly mesenchymal stem cells (WJ-MSC) were isolated and characterized through flow cytometry and trilineage differentiation. For pulmonary differentiation, the cells were seeded in plates coated with natural functional biopolymer matrix as membrane until spheroid formation, and then the spheroids were cultured with differentiation inductors. The differentiated cells were characterized using immunocytochemistry and RT-PCR, confirming the presence of alveolar type I and II, ciliated, and goblet cells. Then, 3D bioprinting was performed with a sodium alginate and gelatin bioink in an extrusion-based 3D printer. The 3D structure was analyzed, confirming cell viability with a live/dead assay and the expression of lung markers with immunocytochemistry. The results showed that the differentiation of WJ-MSC into lung cells was successful, as well as the bioprinting of these cells in a 3D structure, a promising alternative for in vitro drug testing.


Subject(s)
Bioprinting , COVID-19 , Wharton Jelly , Humans , COVID-19/metabolism , Cells, Cultured , Cell Differentiation , Printing, Three-Dimensional , Tissue Engineering
5.
Sci Adv ; 8(43): eabq6900, 2022 10 28.
Article in English | MEDLINE | ID: covidwho-2088382

ABSTRACT

Three-dimensional (3D) bioprinting of vascular tissues that are mechanically and functionally comparable to their native counterparts is an unmet challenge. Here, we developed a tough double-network hydrogel (bio)ink for microfluidic (bio)printing of mono- and dual-layered hollow conduits to recreate vein- and artery-like tissues, respectively. The tough hydrogel consisted of energy-dissipative ionically cross-linked alginate and elastic enzyme-cross-linked gelatin. The 3D bioprinted venous and arterial conduits exhibited key functionalities of respective vessels including relevant mechanical properties, perfusability, barrier performance, expressions of specific markers, and susceptibility to severe acute respiratory syndrome coronavirus 2 pseudo-viral infection. Notably, the arterial conduits revealed physiological vasoconstriction and vasodilatation responses. We further explored the feasibility of these conduits for vascular anastomosis. Together, our study presents biofabrication of mechanically and functionally relevant vascular conduits, showcasing their potentials as vascular models for disease studies in vitro and as grafts for vascular surgeries in vivo, possibly serving broad biomedical applications in the future.


Subject(s)
Bioprinting , COVID-19 , Humans , Bioprinting/methods , Hydrogels , Gelatin , Microfluidics , Tissue Engineering/methods , Printing, Three-Dimensional , Alginates , Tissue Scaffolds
6.
Int J Mol Sci ; 23(19)2022 Sep 21.
Article in English | MEDLINE | ID: covidwho-2066117

ABSTRACT

Vascular replacement is one of the most effective tools to solve cardiovascular diseases, but due to the limitations of autologous transplantation, size mismatch, etc., the blood vessels for replacement are often in short supply. The emergence of artificial blood vessels with 3D bioprinting has been expected to solve this problem. Blood vessel prosthesis plays an important role in the field of cardiovascular medical materials. However, a small-diameter blood vessel prosthesis (diameter < 6 mm) is still unable to achieve wide clinical application. In this paper, a response surface analysis was firstly utilized to obtain the relationship between the contact angle and the gelatin/sodium alginate mixed hydrogel solution at different temperatures and mass percentages. Then, the self-developed 3D bioprinter was used to obtain the optimal printing spacing under different conditions through row spacing, printing, and verifying the relationship between the contact angle and the printing thickness. Finally, the relationship between the blood vessel wall thickness and the contact angle was obtained by biofabrication with 3D bioprinting, which can also confirm the controllability of the vascular membrane thickness molding. It lays a foundation for the following study of the small caliber blood vessel printing molding experiment.


Subject(s)
Bioprinting , Blood Substitutes , Alginates , Blood Vessel Prosthesis , Gelatin , Hydrogels/pharmacology , Printing, Three-Dimensional , Tissue Engineering , Tissue Scaffolds
7.
AAPS PharmSciTech ; 23(7): 267, 2022 Sep 26.
Article in English | MEDLINE | ID: covidwho-2054055

ABSTRACT

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.


Subject(s)
COVID-19 , Chitosan , Zein , Alginates , Biocompatible Materials , Humans , Polyesters , Polymers , Printing, Three-Dimensional , Tissue Engineering/methods , Tissue Scaffolds
8.
Eur J Heart Fail ; 24(10): 1778-1791, 2022 Oct.
Article in English | MEDLINE | ID: covidwho-1981658

ABSTRACT

In vitro modelling the complex (patho-) physiological conditions of the heart is a major challenge in cardiovascular research. In recent years, methods based on three-dimensional (3D) cultivation approaches have steadily evolved to overcome the major limitations of conventional adherent two-dimensional (2D) monolayer cultivation. These 3D approaches aim to study, reproduce or modify fundamental native features of the heart such as tissue organization and cardiovascular microenvironment. Therefore, these systems have great potential for (patient-specific) disease research, for the development of new drug screening platforms, and for the use in regenerative and replacement therapy applications. Consequently, continuous improvement and adaptation is required with respect to fundamental limitations such as cardiomyocyte maturation, scalability, heterogeneity, vascularization, and reproduction of native properties. In this review, 2D monolayer culturing and the 3D in vitro systems of cardiac spheroids, organoids, engineered cardiac microtissue and bioprinting as well as the ex vivo technique of myocardial slicing are introduced with their basic concepts, advantages, and limitations. Furthermore, recent advances of various new approaches aiming to extend as well as to optimize these in vitro and ex vivo systems are presented.


Subject(s)
Bioprinting , Heart Failure , Humans , Bioprinting/methods , Organoids , Myocardium , Myocytes, Cardiac , Tissue Engineering/methods
9.
Int J Biol Macromol ; 219: 694-708, 2022 Oct 31.
Article in English | MEDLINE | ID: covidwho-1977349

ABSTRACT

A new biodegradable semi-interpenetrated polymer network (semi-IPN) of two US Food and Drug Administration approved materials, poly(3-hydroxybutyrate-co-3-valerate) (PHBV) and calcium alginate (CA) was engineered to provide an alternative strategy to enhance the poor adhesion properties of CA. The synthesis procedure allows the additional incorporation of 10 % w/w of graphene nanoplatelets (GNPs), which have no cytotoxic effect on human keratinocytes. This quantity of multilayer graphene provides superior antiviral activity to the novel semi-IPN against a surrogate virus of SARS-CoV-2. Adding GNPs hardly affects the water absorption or electrical conductivity of the pure components of CA and PHBV. However, the semi-IPN's electrical conductivity increases dramatically after adding GNP due to molecular rearrangements of the intertwined polymer chains that continuously distribute the GNP nanosheets, This new hydrophilic composite biomaterial film shows great promise for skin biomedical applications, especially those that require antiviral and/or biodegradable electroconductive materials.


Subject(s)
COVID-19 , Graphite , 3-Hydroxybutyric Acid , Alginates , Antiviral Agents/pharmacology , Biocompatible Materials/pharmacology , Cell Adhesion , Graphite/pharmacology , Humans , Hydrogels/pharmacology , Methylgalactosides , Polyesters/pharmacology , SARS-CoV-2 , Tissue Engineering/methods , Valerates , Water
10.
Stem Cell Res Ther ; 13(1): 317, 2022 07 16.
Article in English | MEDLINE | ID: covidwho-1938351

ABSTRACT

One of the severe complications occurring because of the patient's intubation is tracheal stenosis. Its incidence has significantly risen because of the COVID-19 pandemic and tends only to increase. Here, we propose an alternative to the donor trachea and synthetic prostheses-the tracheal equivalent. To form it, we applied the donor trachea samples, which were decellularized, cross-linked, and treated with laser to make wells on their surface, and inoculated them with human gingiva-derived mesenchymal stromal cells. The fabricated construct was assessed in vivo using nude (immunodeficient), immunosuppressed, and normal mice and rabbits. In comparison with the matrix ones, the tracheal equivalent samples demonstrated the thinning of the capsule, the significant vessel ingrowth into surrounding tissues, and the increase in the submucosa resorption. The developed construct was shown to be highly biocompatible and efficient in trachea restoration. These results can facilitate its clinical translation and be a base to design clinical trials.


Subject(s)
COVID-19 , Tissue Engineering , Animals , Humans , Lasers , Mice , Pandemics , Rabbits , Tissue Engineering/methods , Tissue Scaffolds , Trachea
11.
Nat Commun ; 13(1): 3459, 2022 06 16.
Article in English | MEDLINE | ID: covidwho-1921608

ABSTRACT

Newly developed acoustic technologies are playing a transformational role in life science and biomedical applications ranging from the activation and inactivation of mechanosensitive ion channels for fundamental physiological processes to the development of contact-free, precise biofabrication protocols for tissue engineering and large-scale manufacturing of organoids. Here, we provide our perspective on the development of future acoustic technologies and their promise in addressing critical challenges in biomedicine.


Subject(s)
Acoustics , Sound , Delivery of Health Care , Organoids , Tissue Engineering
12.
Am J Transplant ; 22(6): 1507-1508, 2022 06.
Article in English | MEDLINE | ID: covidwho-1878988
13.
Int J Mol Sci ; 23(9)2022 Apr 21.
Article in English | MEDLINE | ID: covidwho-1818149

ABSTRACT

The impact of COVID-19 has rendered medical technology an important factor to maintain social stability and economic increase, where biomedicine has experienced rapid development and played a crucial part in fighting off the pandemic. Conductive hydrogels (CHs) are three-dimensional (3D) structured gels with excellent electrical conductivity and biocompatibility, which are very suitable for biomedical applications. CHs can mimic innate tissue's physical, chemical, and biological properties, which allows them to provide environmental conditions and structural stability for cell growth and serve as efficient delivery substrates for bioactive molecules. The customizability of CHs also allows additional functionality to be designed for different requirements in biomedical applications. This review introduces the basic functional characteristics and materials for preparing CHs and elaborates on their synthetic techniques. The development and applications of CHs in the field of biomedicine are highlighted, including regenerative medicine, artificial organs, biosensors, drug delivery systems, and some other application scenarios. Finally, this review discusses the future applications of CHs in the field of biomedicine. In summary, the current design and development of CHs extend their prospects for functioning as an intelligent and complex system in diverse biomedical applications.


Subject(s)
COVID-19 , Hydrogels , Biocompatible Materials/chemistry , Biocompatible Materials/therapeutic use , Electric Conductivity , Humans , Hydrogels/chemistry , Hydrogels/therapeutic use , Tissue Engineering/methods
14.
Int J Mol Sci ; 23(7)2022 Mar 22.
Article in English | MEDLINE | ID: covidwho-1785725

ABSTRACT

Tumor cells evolve in a complex and heterogeneous environment composed of different cell types and an extracellular matrix. Current 2D culture methods are very limited in their ability to mimic the cancer cell environment. In recent years, various 3D models of cancer cells have been developed, notably in the form of spheroids/organoids, using scaffold or cancer-on-chip devices. However, these models have the disadvantage of not being able to precisely control the organization of multiple cell types in complex architecture and are sometimes not very reproducible in their production, and this is especially true for spheroids. Three-dimensional bioprinting can produce complex, multi-cellular, and reproducible constructs in which the matrix composition and rigidity can be adapted locally or globally to the tumor model studied. For these reasons, 3D bioprinting seems to be the technique of choice to mimic the tumor microenvironment in vivo as closely as possible. In this review, we discuss different 3D-bioprinting technologies, including bioinks and crosslinkers that can be used for in vitro cancer models and the techniques used to study cells grown in hydrogels; finally, we provide some applications of bioprinted cancer models.


Subject(s)
Bioprinting , Neoplasms , Bioprinting/methods , Humans , Hydrogels , Precision Medicine , Printing, Three-Dimensional , Tissue Engineering/methods , Tissue Scaffolds , Tumor Microenvironment
15.
Int J Mol Sci ; 23(6)2022 Mar 09.
Article in English | MEDLINE | ID: covidwho-1765730

ABSTRACT

Hydrogels are hydrophilic polymer materials that provide a wide range of physicochemical properties as well as are highly biocompatible. Biomedical researchers are adapting these materials for the ever-increasing range of design options and potential applications in diagnostics and therapeutics. Along with innovative hydrogel polymer backbone developments, designing polymer additives for these backbones has been a major contributor to the field, especially for expanding the functionality spectrum of hydrogels. For the past decade, researchers invented numerous hydrogel functionalities that emerge from the rational incorporation of additives such as nucleic acids, proteins, cells, and inorganic nanomaterials. Cases of successful commercialization of such functional hydrogels are being reported, thus driving more translational research with hydrogels. Among the many hydrogels, here we reviewed recently reported functional hydrogels incorporated with polymer additives. We focused on those that have potential in translational medicine applications which range from diagnostic sensors as well as assay and drug screening to therapeutic actuators as well as drug delivery and implant. We discussed the growing trend of facile point-of-care diagnostics and integrated smart platforms. Additionally, special emphasis was given to emerging bioinformatics functionalities stemming from the information technology field, such as DNA data storage and anti-counterfeiting strategies. We anticipate that these translational purpose-driven polymer additive research studies will continue to advance the field of functional hydrogel engineering.


Subject(s)
Hydrogels , Nucleic Acids , Biocompatible Materials , Drug Delivery Systems , Hydrogels/chemistry , Polymers , Tissue Engineering
16.
J Dent Res ; 101(9): 1015-1024, 2022 08.
Article in English | MEDLINE | ID: covidwho-1752997

ABSTRACT

Oral tissue regeneration following chronic diseases and injuries is limited by the natural endogenous wound-healing process. Current regenerative approaches implement exogenous systems, including stem cells, scaffolds, growth factors, and plasmid DNA/viral vectors, that induce variable clinical outcomes. An innovative approach that is safe, effective, and inexpensive is needed. The lipid nanoparticle-encapsulated nucleoside-modified messenger RNA (mRNA) platform has proven to be a successful vaccine modality against coronavirus disease 2019, demonstrating safety and high efficacy in humans. The same fundamental technology platform could be applied to facilitate the development of mRNA-based regenerative therapy. While the platform has not yet been studied in the field of oral tissue regeneration, mRNA therapeutics encoding growth factors have been evaluated and demonstrated promising findings in various models of soft and hard tissue regeneration such as myocardial infarction, diabetic wound healing, and calvarial and femoral bone defects. Because restoration of both soft and hard tissues is crucial to oral tissue physiology, this new therapeutic modality may help to overcome challenges associated with the reconstruction of the unique and complex architecture of oral tissues. This review discusses mRNA therapeutics with an emphasis on findings and lessons in different regenerative animal models, and it speculates how we can apply mRNA-based platforms for oral tissue regeneration.


Subject(s)
COVID-19 , Tissue Engineering , Animals , Bone Regeneration/genetics , Humans , Intercellular Signaling Peptides and Proteins , Liposomes , Nanoparticles , RNA, Messenger , Technology , Wound Healing/genetics
17.
Stem Cells Transl Med ; 11(2): 107-113, 2022 Mar 17.
Article in English | MEDLINE | ID: covidwho-1752179

ABSTRACT

Advances in regenerative medicine manufacturing continue to be a priority for achieving the full commercial potential of important breakthrough therapies. Equally important will be the establishment of distribution chains that support the transport of live cells and engineered tissues and organs resulting from these advanced biomanufacturing processes. The importance of a well-managed distribution chain for products requiring specialized handling procedures was highlighted during the COVID-19 pandemic and serves as a reminder of the critical role of logistics and distribution in the success of breakthrough therapies. This perspective article will provide insight into current practices and future considerations for creating global distribution chains that facilitate the successful deployment of regenerative medicine therapies to the vast number of patients that would benefit from them worldwide.


Subject(s)
COVID-19 , Regenerative Medicine , Cell- and Tissue-Based Therapy , Humans , Pandemics , Regenerative Medicine/methods , Tissue Engineering/methods
18.
J Mater Chem B ; 9(47): 9642-9657, 2021 12 08.
Article in English | MEDLINE | ID: covidwho-1684136

ABSTRACT

Cancer is a growing threat to human beings. Traditional treatments for malignant tumors usually involve invasive means to healthy human tissues, such as surgical treatment and chemotherapy. In recent years the use of specific stimulus-responsive materials in combination with some non-contact, non-invasive stimuli can lead to better efficacy and has become an important area of research. It promises to develop personalized treatment systems for four types of physical stimuli: light, ultrasound, magnetic field, and temperature. Nanomaterials that are responsive to these stimuli can be used to enhance drug delivery, cancer treatment, and tissue engineering. This paper reviews the principles of the stimuli mentioned above, their effects on materials, and how they work with nanomaterials. For this aim, we focus on specific applications in controlled drug release, cancer therapy, tissue engineering, and virus detection, with particular reference to recent photothermal, photodynamic, sonodynamic, magnetothermal, radiation, and other types of therapies. It is instructive for the future development of stimulus-responsive nanomaterials for these aspects.


Subject(s)
Antineoplastic Agents/therapeutic use , Delayed-Action Preparations/therapeutic use , Metal Nanoparticles/therapeutic use , Neoplasms/drug therapy , Radiation-Sensitizing Agents/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/radiation effects , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/radiation effects , Humans , Infrared Rays , Magnetic Phenomena , Metal Nanoparticles/chemistry , Metal Nanoparticles/radiation effects , Radiation-Sensitizing Agents/chemistry , Radiation-Sensitizing Agents/radiation effects , SARS-CoV-2/isolation & purification , Temperature , Tissue Engineering/methods , Ultrasonic Waves , Viral Load/methods
19.
Drug Discov Today ; 27(4): 1062-1076, 2022 04.
Article in English | MEDLINE | ID: covidwho-1587950

ABSTRACT

Proposing efficient prophylactic and therapeutic strategies for coronavirus 2019 (COVID-19) requires precise knowledge of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pathogenesis. An array of platforms, including organoids and microfluidic devices, have provided a basis for studies of SARS-CoV-2. Here, we summarize available models as well as novel drug screening approaches, from simple to more advanced platforms. Notably, organoids and microfluidic devices offer promising perspectives for the clinical translation of basic science, such as screening therapeutics candidates. Overall, modifying these advanced micro and macro 3D platforms for disease modeling and combining them with recent advances in drug screening has significant potential for the discovery of novel potent drugs against COVID-19.


Subject(s)
COVID-19 Drug Treatment , Drug Evaluation, Preclinical , Microfluidics , Models, Biological , Organoids , SARS-CoV-2 , Animals , COVID-19/genetics , Gene Editing , Genome , Humans , Tissue Engineering
20.
Molecules ; 26(21)2021 Nov 06.
Article in English | MEDLINE | ID: covidwho-1502470

ABSTRACT

The normal function of the airway epithelium is vital for the host's well-being. Conditions that might compromise the structure and functionality of the airway epithelium include congenital tracheal anomalies, infection, trauma and post-intubation injuries. Recently, the onset of COVID-19 and its complications in managing respiratory failure further intensified the need for tracheal tissue replacement. Thus far, plenty of naturally derived, synthetic or allogeneic materials have been studied for their applicability in tracheal tissue replacement. However, a reliable tracheal replacement material is missing. Therefore, this study used a tissue engineering approach for constructing tracheal tissue. Human respiratory epithelial cells (RECs) were isolated from nasal turbinate, and the cells were incorporated into a calcium chloride-polymerized human blood plasma to form a human tissue respiratory epithelial construct (HTREC). The quality of HTREC in vitro, focusing on the cellular proliferation, differentiation and distribution of the RECs, was examined using histological, gene expression and immunocytochemical analysis. Histological analysis showed a homogenous distribution of RECs within the HTREC, with increased proliferation of the residing RECs within 4 days of investigation. Gene expression analysis revealed a significant increase (p < 0.05) in gene expression level of proliferative and respiratory epithelial-specific markers Ki67 and MUC5B, respectively, within 4 days of investigation. Immunohistochemical analysis also confirmed the expression of Ki67 and MUC5AC markers in residing RECs within the HTREC. The findings show that calcium chloride-polymerized human blood plasma is a suitable material, which supports viability, proliferation and mucin secreting phenotype of RECs, and this suggests that HTREC can be a potential candidate for respiratory epithelial tissue reconstruction.


Subject(s)
Respiratory Mucosa/metabolism , Tissue Engineering/methods , Trachea/transplantation , Cell Differentiation , Cell Proliferation , Epithelial Cells/metabolism , Epithelium/metabolism , Feasibility Studies , Humans , Ki-67 Antigen/analysis , Ki-67 Antigen/genetics , Mucin 5AC/analysis , Mucin 5AC/genetics , Mucous Membrane/metabolism , Primary Cell Culture/methods , Respiratory Mucosa/physiology , Trachea/metabolism , Trachea/physiology
SELECTION OF CITATIONS
SEARCH DETAIL