Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
Am J Transplant ; 22(6): 1507-1508, 2022 06.
Article in English | MEDLINE | ID: covidwho-1878988
9.
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
10.
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
11.
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
12.
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
13.
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
14.
Adv Drug Deliv Rev ; 179: 114002, 2021 12.
Article in English | MEDLINE | ID: covidwho-1465981

ABSTRACT

After thirty years of intensive research shaping and optimizing the technology, the approval of the first mRNA-based formulation by the EMA and FDA in order to stop the COVID-19 pandemic was a breakthrough in mRNA research. The astonishing success of these vaccines have brought the mRNA platform into the spotlight of the scientific community. The remarkable persistence of the groundwork is mainly attributed to the exceptional benefits of mRNA application, including the biological origin, immediate but transitory mechanism of action, non-integrative properties, safe and relatively simple manufacturing as well as the flexibility to produce any desired protein. Based on these advantages, a practical implementation of in vitro transcribed mRNA has been considered in most areas of medicine. In this review, we discuss the key preconditions for the rise of the mRNA in the medical field, including the unique structural and functional features of the mRNA molecule and its vehicles, which are crucial aspects for a production of potent mRNA-based therapeutics. Further, we focus on the utility of mRNA tools particularly in the scope of regenerative medicine, i.e. cell reprogramming approaches or manipulation strategies for targeted tissue restoration. Finally, we highlight the strong clinical potential but also the remaining hurdles to overcome for the mRNA-based regenerative therapy, which is only a few steps away from becoming a reality.


Subject(s)
Cell- and Tissue-Based Therapy/methods , RNA, Messenger/therapeutic use , Regenerative Medicine/trends , Tissue Engineering/methods , Animals , COVID-19 , Humans
15.
Int J Mol Sci ; 22(3)2021 Jan 26.
Article in English | MEDLINE | ID: covidwho-1389389

ABSTRACT

A high-throughput drug screen identifies potentially promising therapeutics for clinical trials. However, limitations that persist in current disease modeling with limited physiological relevancy of human patients skew drug responses, hamper translation of clinical efficacy, and contribute to high clinical attritions. The emergence of induced pluripotent stem cell (iPSC) technology revolutionizes the paradigm of drug discovery. In particular, iPSC-based three-dimensional (3D) tissue engineering that appears as a promising vehicle of in vitro disease modeling provides more sophisticated tissue architectures and micro-environmental cues than a traditional two-dimensional (2D) culture. Here we discuss 3D based organoids/spheroids that construct the advanced modeling with evolved structural complexity, which propels drug discovery by exhibiting more human specific and diverse pathologies that are not perceived in 2D or animal models. We will then focus on various central nerve system (CNS) disease modeling using human iPSCs, leading to uncovering disease pathogenesis that guides the development of therapeutic strategies. Finally, we will address new opportunities of iPSC-assisted drug discovery with multi-disciplinary approaches from bioengineering to Omics technology. Despite technological challenges, iPSC-derived cytoarchitectures through interactions of diverse cell types mimic patients' CNS and serve as a platform for therapeutic development and personalized precision medicine.


Subject(s)
Central Nervous System Diseases/drug therapy , Drug Discovery/methods , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Tissue Engineering/methods , Animals , COVID-19/pathology , Central Nervous System Diseases/pathology , Drug Discovery/instrumentation , Drug Evaluation, Preclinical/instrumentation , Drug Evaluation, Preclinical/methods , Humans , Induced Pluripotent Stem Cells/pathology , Lab-On-A-Chip Devices , Organoids/cytology , Organoids/drug effects , Organoids/pathology , Tissue Engineering/instrumentation , Zika Virus Infection/drug therapy , Zika Virus Infection/pathology , COVID-19 Drug Treatment
16.
Annu Rev Biomed Eng ; 23: 461-491, 2021 07 13.
Article in English | MEDLINE | ID: covidwho-1191179

ABSTRACT

Modeling immunity in vitro has the potential to be a powerful tool for investigating fundamental biological questions, informing therapeutics and vaccines, and providing new insight into disease progression. There are two major elements to immunity that are necessary to model: primary immune tissues and peripheral tissues with immune components. Here, we systematically review progress made along three strategies to modeling immunity: ex vivo cultures, which preserve native tissue structure; microfluidic devices, which constitute a versatile approach to providing physiologically relevant fluid flow and environmental control; and engineered tissues, which provide precise control of the 3D microenvironment and biophysical cues. While many models focus on disease modeling, more primary immune tissue models are necessary to advance the field. Moving forward, we anticipate that the expansion of patient-specific models may inform why immunity varies from patient to patient and allow for the rapid comprehension and treatment of emerging diseases, such as coronavirus disease 2019.


Subject(s)
COVID-19/immunology , Tissue Engineering/methods , Adaptive Immunity , Animals , Biophysics , Humans , Immune System , Immunity, Innate , In Vitro Techniques , Lab-On-A-Chip Devices , Lymphocytes/immunology , Macrophages/immunology , Mice , Microfluidics , SARS-CoV-2 , Thymus Gland/immunology , Tissue Array Analysis
17.
J Mol Cell Cardiol ; 153: 106-110, 2021 04.
Article in English | MEDLINE | ID: covidwho-989436

ABSTRACT

The coronavirus disease 2019 (COVID-19) outbreak caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a global pandemic as declared by World Health Organization (WHO). In the absence of an effective treatment, different drugs with unknown effectiveness, including antimalarial hydroxychloroquine (HCQ), with or without concurrent administration with azithromycin (AZM), have been tested for treating COVID-19 patients with developed pneumonia. However, the efficacy and safety of HCQ and/or AZM have been questioned by recent clinical reports. Direct effects of these drugs on the human heart remain very poorly defined. To better understand the mechanisms of action of HCQ +/- AZM, we employed bioengineered human ventricular cardiac tissue strip (hvCTS) and anisotropic sheet (hvCAS) assays, made with human pluripotent stem cell (hPSC)-derived ventricular cardiomyocytes (hvCMs), which have been designed for measuring cardiac contractility and electrophysiology, respectively. Our hvCTS experiments showed that AZM induced a dose-dependent negative inotropic effect which could be aggravated by HCQ; electrophysiologically, as revealed by the hvCAS platform, AZM prolonged action potentials and induced spiral wave formations. Collectively, our data were consistent with reported clinical risks of HCQ and AZM on QTc prolongation/ventricular arrhythmias and development of heart failure. In conclusion, our study exposed the risks of HCQ/AZM administration while providing mechanistic insights for their toxicity. Our bioengineered human cardiac tissue constructs therefore provide a useful platform for screening cardiac safety and efficacy when developing therapeutics against COVID-19.


Subject(s)
Arrhythmias, Cardiac/pathology , Azithromycin/adverse effects , Chloroquine/adverse effects , Drug-Related Side Effects and Adverse Reactions/pathology , Myocardial Contraction , Myocytes, Cardiac/pathology , Ventricular Function/drug effects , Anti-Bacterial Agents/adverse effects , Antimalarials/adverse effects , Arrhythmias, Cardiac/chemically induced , Drug-Related Side Effects and Adverse Reactions/etiology , Humans , Myocytes, Cardiac/drug effects , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/pathology , Tissue Engineering/methods , COVID-19 Drug Treatment
18.
Artif Organs ; 45(6): 548-558, 2021 Jun.
Article in English | MEDLINE | ID: covidwho-954035

ABSTRACT

The new coronavirus (2019-nCoV) or the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was officially declared by the World Health Organization (WHO) as a pandemic in March 2020. To date, there are no specific antiviral drugs proven to be effective in treating SARS-CoV-2, requiring joint efforts from different research fronts to discover the best route of treatment. The first decisions in drug discovery are based on 2D cell culture using high-throughput screening. In this context, spheroids and organoids emerge as a reliable alternative. Both are scaffold-free 3D engineered constructs that recapitulate key cellular and molecular events of tissue physiology. Different studies have already shown their advantages as a model for different infectious diseases, including SARS-CoV-2 and for drug screening. The use of these 3D engineered tissues as an in vitro model can fill the gap between 2D cell culture and in vivo preclinical assays (animal models) as they could recapitulate the entire viral life cycle. The main objective of this review is to understand spheroid and organoid biology, highlighting their advantages and disadvantages, and how these scaffold-free engineered tissues can contribute to a better comprehension of viral infection by SARS-CoV-2 and to the development of in vitro high-throughput models for drug screening.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Organoids/physiology , Spheroids, Cellular/physiology , Tissue Engineering/methods , Cells, Cultured , Drug Evaluation, Preclinical , Humans , Organoids/virology , SARS-CoV-2 , Spheroids, Cellular/virology , Tissue Scaffolds
19.
Drug Discov Today ; 26(2): 360-383, 2021 02.
Article in English | MEDLINE | ID: covidwho-923306

ABSTRACT

3D printing was once predicted to be the third industrial revolution. Today, the use of 3D printing is found across almost all industries. This article discusses the latest 3D printing applications in the biomedical industry.


Subject(s)
Biomedical Technology/methods , Printing, Three-Dimensional , Animals , Drug Delivery Systems , Equipment Design , Humans , Prostheses and Implants , Tissue Engineering/methods
20.
Int J Mol Sci ; 21(14)2020 Jul 15.
Article in English | MEDLINE | ID: covidwho-910461

ABSTRACT

Stromal vascular fraction (SVF) containing adipose stem cells (ASCs) has been used for many years in regenerative plastic surgery for autologous applications, without any focus on their potential allogenic role. Allogenic SVF transplants could be based on the possibility to use decellularized extracellular matrix (ECM) as a scaffold from a donor then re-cellularized by ASCs of the recipient, in order to develop the advanced therapy medicinal products (ATMP) in fully personalized clinical approaches. A systematic review of this field has been realized in accordance with the Preferred Reporting for Items for Systematic Reviews and Meta-Analyses-Protocols (PRISMA-P) guidelines. Multistep research of the PubMed, Embase, MEDLINE, Pre-MEDLINE, PsycINFO, CINAHL, Clinicaltrials.gov, Scopus database, and Cochrane databases has been conducted to identify articles and investigations on human allogenic ASCs transplant for clinical use. Of the 341 articles identified, 313 were initially assessed for eligibility on the basis of the abstract. Of these, only 29 met all the predetermined criteria for inclusion according to the PICOS (patients, intervention, comparator, outcomes, and study design) approach, and 19 have been included in quantitative synthesis (meta-analysis). Ninety-one percent of the studies previously screened (284 papers) were focused on the in vitro results and pre-clinical experiments. The allogenic use regarded the treatment of perianal fistulas, diabetic foot ulcers, knee osteoarthritis, acute respiratory distress syndrome, refractory rheumatoid arthritis, pediatrics disease, fecal incontinence, ischemic heart disease, autoimmune encephalomyelitis, lateral epicondylitis, and soft tissue defects. The information analyzed suggested the safety and efficacy of allogenic ASCs and ECM transplants without major side effects.


Subject(s)
Adipose Tissue/transplantation , Extracellular Matrix , Mesenchymal Stem Cell Transplantation , Regenerative Medicine , Tissue Scaffolds , Adipose Tissue/cytology , Animals , Extracellular Matrix/ultrastructure , Humans , Mesenchymal Stem Cell Transplantation/adverse effects , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Regenerative Medicine/methods , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Transplantation, Homologous/adverse effects , Transplantation, Homologous/methods
SELECTION OF CITATIONS
SEARCH DETAIL