Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
2.
Cell Stem Cell ; 31(2): 151-152, 2024 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-38306989

RESUMO

3D printing human tissue models derived from stem cells provides an increasingly popular tissue engineering strategy for probing biological questions. Here Yan et al.1 demonstrate how this technology can be used to model mature human neural tissues with functional neural networks in healthy and disease states.


Assuntos
Bioimpressão , Tecido Nervoso , Humanos , Engenharia Tecidual , Células-Tronco , Impressão Tridimensional
3.
J Neuroinflammation ; 21(1): 32, 2024 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-38263227

RESUMO

Parkinson's disease (PD) and Alzheimer's disease (AD) are neurodegenerative disorders caused by the interaction of genetic, environmental, and familial factors. These diseases have distinct pathologies and symptoms that are linked to specific cell populations in the brain. Notably, the immune system has been implicated in both diseases, with a particular focus on the dysfunction of microglia, the brain's resident immune cells, contributing to neuronal loss and exacerbating symptoms. Researchers use models of the neuroimmune system to gain a deeper understanding of the physiological and biological aspects of these neurodegenerative diseases and how they progress. Several in vitro and in vivo models, including 2D cultures and animal models, have been utilized. Recently, advancements have been made in optimizing these existing models and developing 3D models and organ-on-a-chip systems, holding tremendous promise in accurately mimicking the intricate intracellular environment. As a result, these models represent a crucial breakthrough in the transformation of current treatments for PD and AD by offering potential for conducting long-term disease-based modeling for therapeutic testing, reducing reliance on animal models, and significantly improving cell viability compared to conventional 2D models. The application of 3D and organ-on-a-chip models in neurodegenerative disease research marks a prosperous step forward, providing a more realistic representation of the complex interactions within the neuroimmune system. Ultimately, these refined models of the neuroimmune system aim to aid in the quest to combat and mitigate the impact of debilitating neuroimmune diseases on patients and their families.


Assuntos
Doença de Alzheimer , Doenças Neurodegenerativas , Doença de Parkinson , Animais , Humanos , Sistema Imunitário , Microglia
4.
Int J Mol Sci ; 24(17)2023 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-37686446

RESUMO

Tissue engineering for spinal cord injury (SCI) remains a complex and challenging task. Biomaterial scaffolds have been suggested as a potential solution for supporting cell survival and differentiation at the injury site. However, different biomaterials display multiple properties that significantly impact neural tissue at a cellular level. Here, we evaluated the behavior of different cell lines seeded on chitosan (CHI), poly (ε-caprolactone) (PCL), and poly (L-lactic acid) (PLLA) scaffolds. We demonstrated that the surface properties of a material play a crucial role in cell morphology and differentiation. While the direct contact of a polymer with the cells did not cause cytotoxicity or inhibit the spread of neural progenitor cells derived from neurospheres (NPCdn), neonatal rat spinal cord cells (SCC) and NPCdn only attached and matured on PCL and PLLA surfaces. Scanning electron microscopy and computational analysis suggested that cells attached to the material's surface emerged into distinct morphological populations. Flow cytometry revealed a higher differentiation of neural progenitor cells derived from human induced pluripotent stem cells (hiPSC-NPC) into glial cells on all biomaterials. Immunofluorescence assays demonstrated that PCL and PLLA guided neuronal differentiation and network development in SCC. Our data emphasize the importance of selecting appropriate biomaterials for tissue engineering in SCI treatment.


Assuntos
Células-Tronco Pluripotentes Induzidas , Tecido Nervoso , Traumatismos da Medula Espinal , Regeneração da Medula Espinal , Animais , Ratos , Humanos , Materiais Biocompatíveis/farmacologia , Engenharia Tecidual , Traumatismos da Medula Espinal/terapia
5.
Expert Opin Drug Discov ; 18(9): 1043-1059, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37431937

RESUMO

INTRODUCTION: 3D printing, a versatile additive manufacturing technique, has diverse applications ranging from transportation, rapid prototyping, clean energy, and medical devices. AREAS COVERED: The authors focus on how 3D printing technology can enhance the drug discovery process through automating tissue production that enables high-throughput screening of potential drug candidates. They also discuss how the 3D bioprinting process works and what considerations to address when using this technology to generate cell laden constructs for drug screening as well as the outputs from such assays necessary for determining the efficacy of potential drug candidates. They focus on how bioprinting how has been used to generate cardiac, neural, and testis tissue models, focusing on bio-printed 3D organoids. EXPERT OPINION: The next generation of 3D bioprinted organ model holds great promises for the field of medicine. In terms of drug discovery, the incorporation of smart cell culture systems and biosensors into 3D bioprinted models could provide highly detailed and functional organ models for drug screening. By addressing current challenges of vascularization, electrophysiological control, and scalability, researchers can obtain more reliable and accurate data for drug development, reducing the risk of drug failures during clinical trials.


Assuntos
Bioimpressão , Engenharia Tecidual , Humanos , Engenharia Tecidual/métodos , Organoides , Impressão Tridimensional , Avaliação Pré-Clínica de Medicamentos
6.
Bioelectron Med ; 9(1): 10, 2023 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-37221543

RESUMO

BACKGROUND: Alzheimer's disease (AD), a progressive neurodegenerative disorder, is becoming increasingly prevalent as our population ages. It is characterized by the buildup of amyloid beta plaques and neurofibrillary tangles containing hyperphosphorylated-tau. The current treatments for AD do not prevent the long-term progression of the disease and pre-clinical models often do not accurately represent its complexity. Bioprinting combines cells and biomaterials to create 3D structures that replicate the native tissue environment and can be used as a tool in disease modeling or drug screening. METHODS: This work differentiated both healthy and diseased patient-derived human induced pluripotent stems cells (hiPSCs) into neural progenitor cells (NPCs) that were bioprinted using the Aspect RX1 microfluidic printer into dome-shaped constructs. The combination of cells, bioink, and puromorphamine (puro)-releasing microspheres were used to mimic the in vivo environment and direct the differentiation of the NPCs into basal forebrain-resembling cholinergic neurons (BFCN). These tissue models were then characterized for cell viability, immunocytochemistry, and electrophysiology to evaluate their functionality and physiology for use as disease-specific neural models. RESULTS: Tissue models were successfully bioprinted and the cells were viable for analysis after 30- and 45-day cultures. The neuronal and cholinergic markers ß-tubulin III (Tuj1), forkhead box G1 (FOXG1), and choline acetyltransferase (ChAT) were identified as well as the AD markers amyloid beta and tau. Further, immature electrical activity was observed when the cells were excited with potassium chloride and acetylcholine. CONCLUSIONS: This work shows the successful development of bioprinted tissue models incorporating patient derived hiPSCs. Such models can potentially be used as a tool to screen promising drug candidates for treating AD. Further, this model could be used to increase the understanding of AD progression. The use of patient derived cells also shows the potential of this model for use in personalized medicine applications.

7.
Bio Protoc ; 13(9): e4663, 2023 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-37188103

RESUMO

Three-dimensional bioprinting utilizes additive manufacturing processes that combine cells and a bioink to create living tissue models that mimic tissues found in vivo. Stem cells can regenerate and differentiate into specialized cell types, making them valuable for research concerning degenerative diseases and their potential treatments. 3D bioprinting stem cell-derived tissues have an advantage over other cell types because they can be expanded in large quantities and then differentiated to multiple cell types. Using patient-derived stem cells also enables a personalized medicine approach to the study of disease progression. In particular, mesenchymal stem cells (MSC) are an attractive cell type for bioprinting because they are easier to obtain from patients in comparison to pluripotent stem cells, and their robust characteristics make them desirable for bioprinting. Currently, both MSC bioprinting protocols and cell culturing protocols exist separately, but there is a lack of literature that combines the culturing of the cells with the bioprinting process. This protocol aims to bridge that gap by describing the bioprinting process in detail, starting with how to culture cells pre-printing, to 3D bioprinting the cells, and finally to the culturing process post-printing. Here, we outline the process of culturing MSCs to produce cells for 3D bioprinting. We also describe the process of preparing Axolotl Biosciences TissuePrint - High Viscosity (HV) and Low Viscosity (LV) bioink, the incorporation of MSCs to the bioink, setting up the BIO X and the Aspect RX1 bioprinters, and necessary computer-aided design (CAD) files. We also detail the differentiation of 2D and 3D cell cultures of MSC to dopaminergic neurons, including media preparation. We have also included the protocols for viability, immunocytochemistry, electrophysiology, and performing a dopamine enzyme-linked immunosorbent assay (ELISA), along with the statistical analysis. Graphical overview.

8.
Neurobiol Dis ; 181: 106124, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37054899

RESUMO

Frontotemporal dementia (FTD) refers to a group of neurodegenerative disorders that are characterized by pathology predominantly localized to the frontal and temporal lobes. Approximately 40% of FTD cases are familial, and up to 20% of these are caused by heterozygous loss of function mutations in the gene encoding for progranulin (PGRN), GRN. The mechanisms by which loss of PGRN leads to FTD remain incompletely understood. While astrocytes and microglia have long been linked to the neuropathology of FTD due to mutations in GRN (FTD-GRN), a primary mechanistic role of these supporting cells have not been thoroughly addressed. In contrast, mutations in MAPT, another leading cause of familial FTD, greatly alters astrocyte gene expression leading to subsequent non-cell autonomous effects on neurons, suggesting similar mechanisms may be present in FTD-GRN. Here, we utilized human induced pluripotent stem cell (hiPSC)-derived neural tissue carrying a homozygous GRN R493X-/- knock-in mutation to investigate in vitro whether GRN mutant astrocytes have a non-cell autonomous effect on neurons. Using microelectrode array (MEA) analysis, we demonstrate that the development of spiking activity of neurons cultured with GRN R493X-/- astrocytes was significantly delayed compared to cultures with WT astrocytes. Histological analysis of synaptic markers in these cultures showed an increase in GABAergic synaptic markers and a decrease in glutamatergic synaptic markers during this period when activity was delayed. We also demonstrate that this effect may be due in-part to soluble factors. Overall, this work represents one of the first studies investigating astrocyte-induced neuronal pathology in GRN mutant hiPSCs, and supports the hypothesis of astrocyte involvement in the early pathophysiology of FTD.


Assuntos
Demência Frontotemporal , Células-Tronco Pluripotentes Induzidas , Doença de Pick , Humanos , Demência Frontotemporal/patologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Astrócitos/metabolismo , Progranulinas/genética , Neurônios/metabolismo , Mutação , Doença de Pick/metabolismo
9.
Biomater Sci ; 11(10): 3414-3430, 2023 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-37000528

RESUMO

Cancer is characterized by the uncontrolled division of cells, resulting in the formation of tumors. The tumor microenvironment (TME) consists of a variety of cell types present within a heterogeneous extracellular matrix (ECM). Current 2D culture methods for mimicking this microenvironment remain limited due to spatial constraints. Many different types of 3D cancer models have been developed in recent years using spheroids/organoids, biomaterial scaffolds, and cancer-on-chip systems. However, these models cannot precisely control the organization of multiple cell types inside of complex architectures. Bioprinted cancer models can incorporate both stromal and cancer cells inside of 3D constructs to generate custom models of this complex disease. 3D bioprinting can generate complex, multicellular, and reproducible constructs where the matrix composition and rigidity are tailored locally to the tumor. These capabilities make 3D bioprinting an attractive method for reproducing the tumor TME found in vivo. Recent advancements in biomaterial-based bioinks enable the generation of 3D bioprinted cancer models that accurately mimic the TM. Here we discuss recent examples of such 3D-bioprinted cancer models, including those of the lungs, prostate, skin, brain, and colon. We then highlight the advantages of using 3D bioprinting compared to other in vitro modeling techniques and detail its limitations.


Assuntos
Bioimpressão , Neoplasias , Humanos , Bioimpressão/métodos , Neoplasias/metabolismo , Organoides , Matriz Extracelular/metabolismo , Materiais Biocompatíveis/metabolismo , Impressão Tridimensional , Engenharia Tecidual/métodos , Tecidos Suporte , Microambiente Tumoral
10.
Biomater Sci ; 11(10): 3512-3523, 2023 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-36992650

RESUMO

As nucleic acid (NA) technologies continue to revolutionize medicine, new delivery vehicles are needed to effectively transport NA cargoes into cells. Uniform and length-tunable nanofiber micelleplexes have recently shown promise as versatile polymeric delivery vehicles for plasmid DNA, however the effects of several key parameters on micelleplex transfection and stability remain unknown. In this work, we compare poly(fluorenetrimethylenecarbonate)-b-poly(2-(dimethylamino)ethyl methacrylate) (PFTMC-b-PDMAEMA) nanofiber micelleplexes to nanosphere micelleplexes and PDMAEMA polyplexes, examining the effects of complexation buffer, the temporal and serum stability of nanofiber micelleplexes, as well as the effects of cell density, cell type, and polymer DPn upon transfection efficiency and cell viability. These studies are vital for understanding the formation and biological activity of micelleplexes in more detail and should inform the future design of more advanced polymeric NA delivery systems.


Assuntos
Nanofibras , Metacrilatos , DNA , Polímeros , Nylons , Transfecção
11.
Adv Biol (Weinh) ; 7(7): e2200322, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36895072

RESUMO

Infertility affects 10-15% of couples, with half attributed to male factors. An improved understanding of the cell-type-specific dysfunction contributing to male infertility is needed to improve available therapies; however, human testicular tissues are difficult to obtain for research purposes. To overcome this, researchers have begun to use human induced pluripotent stem cells (hiPSCs) to generate various testis-specific cell types in vitro. Peritubular myoid cells (PTMs) are one such testicular cell type that serves a critical role in the human testis niche but, to date, have not been derived from hiPSCs. This study set forth to generate a molecular-based differentiation method for deriving PTMs from hiPSCs, mirroring in vivo patterning factors. Whole transcriptome profiling and quantitative polymerase chain reaction (qPCR) show that this differentiation method is sufficient to derive cells with PTM-like transcriptomes, including upregulation of hallmark PTM functional genes, secreted growth and matrix factors, smooth muscle, integrins, receptors, and antioxidants. Hierarchical clustering shows that they acquire transcriptomes similar to primary isolated PTMs, and immunostaining shows the acquisition of a smooth muscle phenotype. Overall, these hiPSC-PTMs will allow in vitro study of patient-specific PTM development and function in spermatogenesis and infertility.


Assuntos
Células-Tronco Pluripotentes Induzidas , Infertilidade Masculina , Humanos , Masculino , Testículo/metabolismo , Espermatogênese/genética , Diferenciação Celular/genética , Infertilidade Masculina/metabolismo
13.
Environ Adv ; 9: 100310, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36321068

RESUMO

We report wastewater surveillance of the spread of SARS-CoV-2 based upon 24-h composite influent samples taken weekly from four wastewater treatment plants (WWTP) on Vancouver Island, BC, Canada between January 3, 2021 and July 10, 2021. Samples were analyzed by reverse transcription quantitative polymerase chain reaction targeting the N1 and N2 gene fragments of SARS-CoV-2 and a region of the replication associate protein of the pepper mottle mosaic virus (PMMoV) serving as endemic control. Only a small proportion of samples had quantifiable levels of N1 or N2. Overall case rates are weakly correlated with the concentration (gene copies/L) and with the flux of viral material influent to the WWTP (gene copies/day); the latter accounts for influent flow variations. Poisson multimodal rank correlation accounts for differences between the four WWTP and shows a significant correlation with a significant positive intercept. Receiver operator characteristics (ROC) analysis confirms a cut-off of cases based on amplified/not-amplified experimental data. At the optimal cut point of 19 (N1) or 17 (N2) cases/week/100,000 the sensitivity and specificity is about 75% for N1 and 67% for N2.

14.
J Am Chem Soc ; 144(43): 19799-19812, 2022 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-36260789

RESUMO

Micelleplexes show great promise as effective polymeric delivery systems for nucleic acids. Although studies have shown that spherical micelleplexes can exhibit superior cellular transfection to polyplexes, to date there has been no report on the effects of micelleplex morphology on cellular transfection. In this work, we prepared precision, length-tunable poly(fluorenetrimethylenecarbonate)-b-poly(2-(dimethylamino)ethyl methacrylate) (PFTMC16-b-PDMAEMA131) nanofiber micelleplexes and compared their properties and transfection activity to those of the equivalent nanosphere micelleplexes and polyplexes. We studied the DNA complexation process in detail via a range of techniques including cryo-transmission electron microscopy, atomic force microscopy, dynamic light scattering, and ζ-potential measurements, thereby examining how nanofiber micelleplexes form, as well the key differences that exist compared to nanosphere micelleplexes and polyplexes in terms of DNA loading and colloidal stability. The effects of particle morphology and nanofiber length on the transfection and cell viability of U-87 MG glioblastoma cells with a luciferase plasmid were explored, revealing that short nanofiber micelleplexes (length < ca. 100 nm) were the most effective delivery vehicle examined, outperforming nanosphere micelleplexes, polyplexes, and longer nanofiber micelleplexes as well as the Lipofectamine 2000 control. This study highlights the potential importance of 1D micelleplex morphologies for achieving optimal transfection activity and provides a fundamental platform for the future development of more effective polymeric nucleic acid delivery vehicles.


Assuntos
Nanofibras , Ácidos Nucleicos , Micelas , Transfecção , Polímeros , DNA
15.
STAR Protoc ; 3(2): 101348, 2022 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-35509974

RESUMO

3D bioprinting-a type of additive manufacturing-can create 3D tissue constructs resembling in vivo tissues. Here, we present a protocol for 3D printing neural tissues using Axolotl Biosciences' fibrin-based bioink and the CELLINK BIO X bioprinter with a pneumatic printhead. This workflow can be applied to printing 3D tissue models using a variety of cell lines and any chemically crosslinked bioink. These 3D-printed tissue models can be used for applications such as drug screening and disease modeling in vitro.


Assuntos
Bioimpressão , Tecido Nervoso , Impressão Tridimensional , Engenharia Tecidual/métodos , Tecidos Suporte
16.
F S Sci ; 3(2): 130-139, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35560010

RESUMO

OBJECTIVE: To study the feasibility and spermatogenic potential of 3-dimensional (3D) bioprinting personalized human testicular cells derived from a patient with nonobstructive azoospermia (NOA). DESIGN: A human testicular biopsy from a single donor with NOA was dissociated into single cells, expanded in vitro, and 3D bioprinted into tubular structures akin to the seminiferous tubule using AGC-10 bioink and an RX1 bioprinter with a CENTRA coaxial microfluidic printhead from Aspect Biosystems. Three-dimensional organoid cultures were used as a nonbioprinted in vitro control. SETTING: Academic medical center. PATIENT(S): A 31-year-old man with NOA with testis biopsy demonstrating Sertoli cell-only syndrome. INTERVENTION(S): Three-dimensional bioprinting and in vitro culturing of patient-derived testis cells. MAIN OUTCOME MEASURE(S): Cellular viability after printing was determined, along with the expression of phenotypic and spermatogenic functional genetic markers after 12 days of in vitro culture. RESULT(S): Testicular cultures were expandable in vitro and generated sufficiently large numbers for 3D bioprinting at 35 million cells per mL of bioink. Viability 24 hours after printing was determined to be 93.4% ± 2.4%. Immunofluorescence staining for the phenotype markers SRY-Box transcription factor 9, insulin-like 3, actin alpha 2 smooth muscle, and synaptonemal complex protein 3 after 12 days was positive, confirming the presence of Sertoli, Leydig, peritubular myoid, and meiotic germ cells. Reverse transcription qualitative polymerase chain reaction analysis showed that after 12 days in spermatogenic media, the bioprints substantially up-regulated spermatogenic gene expression on par with nonbioprinted controls and showed a particularly significant improvement in genes involved in spermatogonial stem cell maintenance: inhibitor of deoxyribonucleic acid binding 4 by 365-fold; fibroblast growth factor 3 by 94,152-fold; stem cell growth factor receptor KIT by twofold; stimulated by retinoic acid 8 by 125-fold; deleted in azoospermia-like by 114-fold; synaptonemal complex protein 3 by sevenfold; zona pellucida binding protein by twofold; transition protein 1 by 2,908-fold; and protamine 2 by 11-fold. CONCLUSION(S): This study demonstrates for the first time the feasibility of 3D bioprinting adult human testicular cells. We show that the bioprinting process is compatible with high testicular cell viability and without loss of the main somatic phenotypes within the testis tissue. We demonstrate an increase in germ cell markers in the 3D bioprinted tubules after 12 days of in vitro culture. This platform may carry future potential for disease modeling and regenerative opportunities in a personalized medicine framework.


Assuntos
Azoospermia , Testículo , Azoospermia/genética , Humanos , Masculino , Células de Sertoli , Espermatogênese/genética
17.
Mater Today Bio ; 13: 100221, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35243296

RESUMO

Ischemia occurs when blood flow is reduced or restricted, leading to a lack of oxygen and nutrient supply and removal of metabolites in a body part. Critical limb ischemia (CLI) is a severe clinical manifestation of peripheral arterial disease. Atherosclerosis serves as the main cause of CLI, which arises from the deposition of lipids in the artery wall, forming atheroma and causing inflammation. Although several therapies exist for the treatment of CLI, pharmacotherapy still has low efficacy, and vascular surgery often cannot be performed due to the pathophysiological heterogeneity of each patient. Gene and cell therapies have emerged as alternative treatments for the treatment of CLI by promoting angiogenesis. However, the delivery of autologous, heterologous or genetically modified cells into the ischemic tissue remains challenging, as these cells can die at the injection site and/or leak into other tissues. The encapsulation of these cells within hydrogels for local delivery is probably one of the promising options today. Hydrogels, three-dimensional (3D) cross-linked polymer networks, enable manipulation of physical and chemical properties to mimic the extracellular matrix. Thus, specific biostructures can be developed by adjusting prepolymer properties and encapsulation process variables, such as viscosity and flow rate of fluids, depending on the final biomedical application. Electrostatic droplet extrusion, micromolding, microfluidics, and 3D printing have been the most commonly used technologies for cell encapsulation due to their versatility in producing different hydrogel-based systems (e.g., microgels, fibers, vascularized architectures and perfusable single vessels) with great potential to treat ischemic diseases. This review discusses the cell encapsulation technologies associated with hydrogels which are currently used for advanced therapies applied to limb ischemia, describing their principles, advantages, disadvantages, potentials, and innovative therapeutic ideas.

18.
Biomolecules ; 12(1)2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-35053289

RESUMO

3D bioprinting has tremendous potential to revolutionize the field of regenerative medicine by automating the process of tissue engineering. A significant number of new and advanced bioprinting technologies have been developed in recent years, enabling the generation of increasingly accurate models of human tissues both in the healthy and diseased state. Accordingly, this technology has generated a demand for smart bioinks that can enable the rapid and efficient generation of human bioprinted tissues that accurately recapitulate the properties of the same tissue found in vivo. Here, we define smart bioinks as those that provide controlled release of factors in response to stimuli or combine multiple materials to yield novel properties for the bioprinting of human tissues. This perspective piece reviews the existing literature and examines the potential for the incorporation of micro and nanotechnologies into bioinks to enhance their properties. It also discusses avenues for future work in this cutting-edge field.


Assuntos
Bioimpressão , Tecidos Suporte , Humanos , Impressão Tridimensional , Medicina Regenerativa , Engenharia Tecidual
19.
Rev Neurosci ; 33(3): 257-268, 2022 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-34388333

RESUMO

The COVID-19 pandemic has affected millions of people worldwide. While coronaviruses typically have low rates of neurotropic effects, the massive transmission of SARS-CoV-2 suggests that a substantial population will suffer from potential SARS-CoV-2-related neurological disorders. The rapid and recent emergence of SARS-CoV-2 means little research exists on its potential neurological effects. Here we analyze the effects of similar viruses to provide insight into the potential effects of SARS-CoV-2 on the nervous system and beyond. Seven coronavirus strains (HCoV-OC43, HCoV-HKU1, HCoV-229E, HCoV-NL63, SARS-CoV, MERS-CoV, SARS-CoV-2) can infect humans. Many of these strains cause neurological effects, such as headaches, dizziness, strokes, seizures, and critical illness polyneuropathy/myopathy. Certain studies have also linked coronaviruses with multiple sclerosis and extensive central nervous system injuries. Reviewing these studies provides insight into the anticipated effects for patients with SARS-CoV-2. This review will first describe the effects of other coronaviruses that have caused severe disease (SARS-CoV, MERS-CoV) on the nervous system, as well as their proposed origins, non-neurological effects, and neurological infection mechanisms. It will then discuss what is known about SARS-CoV-2 in these areas with reference to the aforementioned viruses, with the goal of providing a holistic picture of SARS-CoV-2.


Assuntos
COVID-19 , Coronavirus Humano OC43 , Humanos , Sistema Nervoso , Pandemias , SARS-CoV-2
20.
Curr Protoc ; 1(12): e331, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34919351

RESUMO

The ability of stem cells to differentiate into specialized cells make them a valuable tool for therapeutic applications. 3D bioprinting, a subset of additive manufacturing, uses bioinks composed of cells and biomaterials to create living tissues. The use of bioactive factors like small molecules and proteins can promote stem cell differentiation into the desired cell phenotypes for tissue regeneration. Small molecules can accelerate the process of regeneration in tissue engineering, maintain bioactivity in a biological environment, and minimize the costs associated with this process. Additionally, they can be encapsulated in specialized drug-delivery devices called microspheres for controlled release. Microspheres are small (1-1000 µm) spherical particles usually made from biodegradable and biocompatible polymers that can be loaded with drugs and other bioactive components. They can then be integrated into stem-cell-laden bioinks used to form bioprinted tissues, where they will release the encapsulated drug and promote differentiation of stem cells into the desired mature cell type. Microspheres can be widely used to encapsulate a broad range of therapeutic agents, including hydrophilic and hydrophobic small molecule drugs, DNA, and proteins. The release of encapsulated molecules occurs through degradation and erosion of the polymer matrix. This article provides detailed protocols for fabricating and sterilizing drug-releasing microspheres made from poly-ε-caprolactone, a promising biodegradable polymer often used for controlled drug delivery due to its biocompatibility and biodegradation kinetics. Additional protocols describe characterization of the loading and size of microspheres as well as incorporation of microspheres into a fibrin-based bioink for 3D bioprinting. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Fabrication of drug-releasing PCL microspheres Support Protocol 1: Preparation of microspheres for determination of encapsulation efficiency by HPLC Support Protocol 2: Preparation of microspheres for SEM analysis Basic Protocol 2: Incorporation of microspheres into fibrin-based bioink.


Assuntos
Preparações Farmacêuticas , Diferenciação Celular , Microesferas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...