Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
J Vis Exp ; (203)2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38314824

ABSTRACT

Calcium signaling is an integral regulator of nearly every tissue. Within the intestinal epithelium, calcium is involved in the regulation of secretory activity, actin dynamics, inflammatory responses, stem cell proliferation, and many other uncharacterized cellular functions. As such, mapping calcium signaling dynamics within the intestinal epithelium can provide insight into homeostatic cellular processes and unveil unique responses to various stimuli. Human intestinal organoids (HIOs) are a high-throughput, human-derived model to study the intestinal epithelium and thus represent a useful system to investigate calcium dynamics. This paper describes a protocol to stably transduce HIOs with genetically encoded calcium indicators (GECIs), perform live fluorescence microscopy, and analyze imaging data to meaningfully characterize calcium signals. As a representative example, 3-dimensional HIOs were transduced with lentivirus to stably express GCaMP6s, a green fluorescent protein-based cytosolic GECI. The engineered HIOs were then dispersed into a single-cell suspension and seeded as monolayers. After differentiation, the HIO monolayers were infected with rotavirus and/or treated with drugs known to stimulate a calcium response. An epifluorescence microscope fitted with a temperature-controlled, humidified live-imaging chamber allowed for long-term imaging of infected or drug-treated monolayers. Following imaging, acquired images were analyzed using the freely available analysis software, ImageJ. Overall, this work establishes an adaptable pipeline for characterizing cellular signaling in HIOs.


Subject(s)
Calcium , Intestines , Humans , Calcium/analysis , Intestinal Mucosa/chemistry , Organoids/chemistry , Microscopy, Fluorescence/methods
2.
J Mater Chem B ; 11(31): 7442-7453, 2023 08 09.
Article in English | MEDLINE | ID: mdl-37439116

ABSTRACT

The mechanical elasticity or stiffness of the ECM modulates YAP activity to regulate the differentiation of stem cells during the development and defect regeneration of cartilage tissue. However, the understanding of the scaffold-associated mechanobiology during the initiation of chondrogenesis and hyaline cartilaginous phenotype maintenance remains unclear. In order to elucidate such mechanisms to promote articular cartilage repair by producing more hyaline cartilage, we identify the relationship between YAP subcellular localization and variation of the cartilage structure and organization during the early postnatal explosive growth in incipient articular cartilage. Next, we prepared a decellularized cartilage scaffold with different stiffness (2-33 kPa) to investigate the effect of scaffold stiffness on the formation of hyaline cartilage by mesenchymal stem cells and the change of YAP activity. Furthermore, we simulated the decrease of cellular YAP activity during postnatal cartilage development by inhibiting YAP activity with verteporfin, and realized that the timing of drug incorporation was critical to regulate the differentiation of MSCs to hyaline chondrocytes and inhibit their hypertrophy and fibrosis. On this basis, we constructed hyaline cartilage organoids by decellularized matrix scaffolds. Collectively, the results herein demonstrate that YAP plays a critical role during in vitro chondrogenic differentiation which is tightly regulated by biochemical and mechanical regulation.


Subject(s)
Cartilage, Articular , Mesenchymal Stem Cells , Animals , Swine , Cells, Cultured , Mesenchymal Stem Cells/chemistry , Mesenchymal Stem Cells/metabolism , Organoids/chemistry , Verteporfin/chemistry , Biomimetic Materials/chemistry , Cartilage, Articular/chemistry , YAP-Signaling Proteins/chemistry , YAP-Signaling Proteins/metabolism
3.
Lab Chip ; 23(12): 2693-2709, 2023 06 13.
Article in English | MEDLINE | ID: mdl-37256563

ABSTRACT

Modelling the human brain in vitro has been extremely challenging due to the brain's intricate cellular composition and specific structural architecture. The recent emergence of brain organoids that recapitulate many key features of human brain development has thus piqued the interest of many to further develop and apply this in vitro model for various physiological and pathological investigations. Despite ongoing efforts, the existing brain organoids demonstrate several limitations, such as the lack of a functional human vasculature with perfusion capability. Microfluidics is suited to enhance such brain organoid models by enabling vascular perfusion and a curated blood-brain barrier microenvironment. In this review, we first provide an introduction to in vivo human brain development and present the state-of-the-art in vitro human brain models. We further elaborate on different strategies to improve the vascularized human brain organoid microenvironment using microfluidic devices, while discussing the current obstacles and future directions in this field.


Subject(s)
Brain , Organoids , Humans , Organoids/chemistry , Microfluidics
4.
ACS Sens ; 8(2): 712-720, 2023 02 24.
Article in English | MEDLINE | ID: mdl-36749605

ABSTRACT

Common symptoms of food intolerance are caused by chemical components within food that have a pharmacological activity to alter the motility of the gastrointestinal tract. Food intolerance is difficult to diagnose as it requires a long-term process of eliminating foods that are responsible for gastrointestinal symptoms. Enterochromaffin (EC) cells are key intestinal epithelium cells that respond to luminal chemical stimulants by releasing 5-HT. Changes in 5-HT levels have been shown to directly alter the motility of the intestinal tract. Therefore, a rapid approach for monitoring the impact of chemicals in food components on 5-HT levels can provide a personalized insight into food intolerance and help stratify diets. Within this study, we developed a three-dimensional (3D)-printed electrochemical multiwell plate to determine changes in 5-HT levels from intestinal organoids that were exposed to varying chemical components found in food. The carbon black/poly-lactic acid (CB/PLA) electrodes had a linear range in physiological concentrations of 5-HT (0.1-2 µM) with a limit of detection of 0.07 µM. The electrodes were stable for monitoring 5-HT overflow from intestinal organoids. Using the electrochemical multiwell plate containing intestinal organoids, increases in 5-HT were observed in the presence of 0.1 mM cinnamaldehyde and 10 mM quercetin but reduction in 5-HT levels was observed in 1 mM sorbitol when compared to control. These changes in the presence of chemicals commonly found in food were verified with ex vivo ileum tissue measurements using chromatography and amperometry with boron-doped diamond electrodes. Overall, our 3D electrochemical multiwell plate measurements with intestinal organoids highlight an approach that can be a high-throughput platform technology for rapid screening of food intolerance to provide personalized nutritional diet.


Subject(s)
Food Intolerance , Serotonin , Humans , Serotonin/analysis , Ileum/chemistry , Intestinal Mucosa/chemistry , Organoids/chemistry
5.
Sci Total Environ ; 806(Pt 1): 150328, 2022 Feb 01.
Article in English | MEDLINE | ID: mdl-34571217

ABSTRACT

Microplastic particles (MP) has been detected in the environment widespread. Human beings are inevitably exposed to MP via multiple routines. However, the hazard identifications, as direct evidence of exposure and health risk, have not been fully characterized in human beings. Many studies suggest the liver is a potential target organ, but currently no study regarding the MP on human liver has been reported. In this study, we used a novel in vitro 3D model, the liver organoids (LOs) generated from human pluripotent stem cells, as an alternative model to the human liver, to explore the adverse biological effect of 1 µm polystyrene-MP (PS-MP) microbeads applying a non-static exposure approach. When the LOs were exposed to 0.25, 2.5 and 25 µg/mL PS-MP (the lowest one was relevant to the environmental concentrations, calculated to be 102 ± 7 items/mL). The potential mechanisms of PS-MP induced hepatotoxicity and lipotoxicity, in aspects of cytotoxicity, levels of key molecular markers, ATP production, alteration in lipid metabolism, ROS generation, oxidative stress and inflammation response, were determined. Specifically, it has been firstly observed that PS-MP could increase the expression of hepatic HNF4A and CYP2E1. Based on these findings, the potential adverse outcome pathways (AOPs) relevant to PS-MP were proposed, and the potential risks of PS-MP on liver steatosis, fibrosis and cancer were implicated. The combined application of novel LOs model and AOPs framework provides a new insight into the risk assessment of MP. Further studies are anticipated to validate the hepatotoxic molecular mechanism of PS-MP based on HNF4A or CYP2E1, and to investigate the MP-induced physical damage and its relationship to hepatic adverse effect for human beings. CAPSULE: Microplastics cause hepatotoxicity and disrupt lipid metabolism in the human pluripotent stem cells-derived liver organoids, providing evidence for human implication.


Subject(s)
Chemical and Drug Induced Liver Injury , Water Pollutants, Chemical , Humans , Lipid Metabolism , Microplastics , Organoids/chemistry , Plastics/toxicity , Polystyrenes/toxicity , Water Pollutants, Chemical/analysis , Water Pollutants, Chemical/toxicity
6.
Nat Commun ; 12(1): 7159, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34887382

ABSTRACT

The most successful genetically encoded calcium indicators (GECIs) employ an intensity or ratiometric readout. Despite a large calcium-dependent change in fluorescence intensity, the quantification of calcium concentrations with GECIs is problematic, which is further complicated by the sensitivity of all GECIs to changes in the pH in the biological range. Here, we report on a sensing strategy in which a conformational change directly modifies the fluorescence quantum yield and fluorescence lifetime of a circular permutated turquoise fluorescent protein. The fluorescence lifetime is an absolute parameter that enables straightforward quantification, eliminating intensity-related artifacts. An engineering strategy that optimizes lifetime contrast led to a biosensor that shows a 3-fold change in the calcium-dependent quantum yield and a fluorescence lifetime change of 1.3 ns. We dub the biosensor Turquoise Calcium Fluorescence LIfeTime Sensor (Tq-Ca-FLITS). The response of the calcium sensor is insensitive to pH between 6.2-9. As a result, Tq-Ca-FLITS enables robust measurements of intracellular calcium concentrations by fluorescence lifetime imaging. We demonstrate quantitative imaging of calcium concentrations with the turquoise GECI in single endothelial cells and human-derived organoids.


Subject(s)
Biosensing Techniques/methods , Calcium/analysis , Endothelial Cells/metabolism , Luminescent Proteins/chemistry , Biosensing Techniques/instrumentation , Calcium/metabolism , Endothelial Cells/chemistry , Fluorescence , HeLa Cells , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Organoids/chemistry , Organoids/metabolism
7.
Clin Transl Med ; 11(12): e566, 2021 12.
Article in English | MEDLINE | ID: mdl-34954911

ABSTRACT

The well-established 3D organoid culture method enabled efficient expansion of cholangiocyte-like cells from intrahepatic (IHBD) and extrahepatic bile duct (EHBD) tissue biopsies. The extensive expansion capacity of these organoids enables various applications, from cholangiocyte disease modelling to bile duct tissue engineering. Recent research demonstrated the feasibility of culturing cholangiocyte organoids from bile, which was minimal-invasive collected via endoscopic retrograde pancreaticography (ERCP). However, a detailed analysis of these bile cholangiocyte organoids (BCOs) and the cellular region of origin was not yet demonstrated. In this study, we characterize BCOs and mirror them to the already established organoids initiated from IHBD- and EHBD-tissue. We demonstrate successful organoid-initiation from extrahepatic bile collected from gallbladder after resection and by ERCP or percutaneous transhepatic cholangiopathy from a variety of patients. BCOs initiated from these three sources of bile all show features similar to in vivo cholangiocytes. The regional-specific characteristics of the BCOs are reflected by the exclusive expression of regional common bile duct genes (HOXB2 and HOXB3) by ERCP-derived BCOs and gallbladder-derived BCOs expressing gallbladder-specific genes. Moreover, BCOs have limited hepatocyte-fate differentiation potential compared to intrahepatic cholangiocyte organoids. These results indicate that organoid-initiating cells in bile are likely of local (extrahepatic) origin and are not of intrahepatic origin. Regarding the functionality of organoid initiating cells in bile, we demonstrate that BCOs efficiently repopulate decellularized EHBD scaffolds and restore the monolayer of cholangiocyte-like cells in vitro. Bile samples obtained through minimally invasive procedures provide a safe and effective alternative source of cholangiocyte organoids. The shedding of (organoid-initiating) cholangiocytes in bile provides a convenient source of organoids for regenerative medicine.


Subject(s)
Bile Acids and Salts/genetics , Bile Ducts/chemistry , Organoids/chemistry , Phenotype , Adolescent , Adult , Aged , Bile Ducts/physiopathology , Female , Humans , Male , Middle Aged , Organoids/metabolism
8.
Fluids Barriers CNS ; 18(1): 43, 2021 Sep 20.
Article in English | MEDLINE | ID: mdl-34544422

ABSTRACT

BACKGROUND: The pathways that control protein transport across the blood-brain barrier (BBB) remain poorly characterized. Despite great advances in recapitulating the human BBB in vitro, current models are not suitable for systematic analysis of the molecular mechanisms of antibody transport. The gaps in our mechanistic understanding of antibody transcytosis hinder new therapeutic delivery strategy development. METHODS: We applied a novel bioengineering approach to generate human BBB organoids by the self-assembly of astrocytes, pericytes and brain endothelial cells with unprecedented throughput and reproducibility using micro patterned hydrogels. We designed a semi-automated and scalable imaging assay to measure receptor-mediated transcytosis of antibodies. Finally, we developed a workflow to use CRISPR/Cas9 gene editing in BBB organoid arrays to knock out regulators of endocytosis specifically in brain endothelial cells in order to dissect the molecular mechanisms of receptor-mediated transcytosis. RESULTS: BBB organoid arrays allowed the simultaneous growth of more than 3000 homogenous organoids per individual experiment in a highly reproducible manner. BBB organoid arrays showed low permeability to macromolecules and prevented transport of human non-targeting antibodies. In contrast, a monovalent antibody targeting the human transferrin receptor underwent dose- and time-dependent transcytosis in organoids. Using CRISPR/Cas9 gene editing in BBB organoid arrays, we showed that clathrin, but not caveolin, is required for transferrin receptor-dependent transcytosis. CONCLUSIONS: Human BBB organoid arrays are a robust high-throughput platform that can be used to discover new mechanisms of receptor-mediated antibody transcytosis. The implementation of this platform during early stages of drug discovery can accelerate the development of new brain delivery technologies.


Subject(s)
Antibodies/metabolism , Bioengineering/methods , Blood-Brain Barrier/metabolism , Organoids/metabolism , Receptors, Transferrin/metabolism , Transcytosis/physiology , Animals , Antibodies/analysis , Astrocytes/chemistry , Astrocytes/metabolism , Blood-Brain Barrier/chemistry , Blood-Brain Barrier/cytology , Cells, Cultured , Coculture Techniques , Endothelial Cells/chemistry , Endothelial Cells/metabolism , Humans , Organoids/chemistry , Organoids/cytology , Pericytes/chemistry , Pericytes/metabolism , Receptors, Transferrin/analysis
9.
Adv Mater ; 33(46): e2005944, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34270839

ABSTRACT

Cell-based living materials, including single cells, cell-laden fibers, cell sheets, organoids, and organs, have attracted intensive interests owing to their widespread applications in cancer therapy, regenerative medicine, drug development, and so on. Significant progress in materials, microfabrication, and cell biology have promoted the development of numerous promising microfluidic platforms for programming these cell-based living materials with a high-throughput, scalable, and efficient manner. In this review, the recent progress of novel microfluidic platforms for programming cell-based living materials is presented. First, the unique features, categories, and materials and related fabrication methods of microfluidic platforms are briefly introduced. From the viewpoint of the design principles of the microfluidic platforms, the recent significant advances of programming single cells, cell-laden fibers, cell sheets, organoids, and organs in turns are then highlighted. Last, by providing personal perspectives on challenges and future trends, this review aims to motivate researchers from the fields of materials and engineering to work together with biologists and physicians to promote the development of cell-based living materials for human healthcare-related applications.


Subject(s)
Microfluidics/methods , Animals , Biocompatible Materials/chemistry , Bioprinting , Cellular Reprogramming , Humans , Hydrogels/chemistry , Microfluidics/instrumentation , Nanostructures/chemistry , Organoids/chemistry , Regenerative Medicine
10.
Mikrochim Acta ; 188(6): 203, 2021 05 27.
Article in English | MEDLINE | ID: mdl-34043106

ABSTRACT

Molecularly imprinted polymer (MIP)-based electrochemical sensors for the protein α-synuclein (a marker for Parkinson's disease) were developed using a peptide epitope from the protein. MIPs doped with various concentrations and species of transition metal dichalcogenides (TMDs) to enhance conductivity were electropolymerized with and without template molecules. The current during the electropolymerization was compared with that associated with the electrochemical response (at 0.24~0.29 V vs. ref. electrode) to target peptide molecules in the finished sensor. We found that this relationship can aid in the rational design of conductive MIPs for the recognition of biomarkers in biological fluids. The sensing range and limit of detection of TMD-doped imprinted poly(AN-co-MSAN)-coated electrodes were 0.001-100 pg/mL and 0.5 fg/mL (SNR = 3), respectively. To show the potential applicability of the MIP electrochemical sensor, cell culture medium from PD patient-specific midbrain organoids generated from induced pluripotent stem cells was analyzed. α-Synuclein levels were found to be significantly reduced in the organoids from PD patients, compared to those generated from age-matched controls. The relative standard deviation and recovery are less than 5% and 95-115%, respectively. Preparation of TMD-doped α-synuclein (SNCA) peptide-imprinted poly(AN-co-MSAN)-coated electrodes.


Subject(s)
Disulfides/chemistry , Molecularly Imprinted Polymers/chemistry , Molybdenum/chemistry , Sulfides/chemistry , Tungsten Compounds/chemistry , alpha-Synuclein/analysis , Electrochemical Techniques/methods , Humans , Limit of Detection , Mesencephalon/chemistry , Organoids/chemistry , Parkinson Disease/diagnosis , Peptide Fragments/chemistry , alpha-Synuclein/chemistry
11.
Methods Mol Biol ; 2299: 435-445, 2021.
Article in English | MEDLINE | ID: mdl-34028759

ABSTRACT

The mechanisms of kidney injury and fibrosis can now be studied using kidney organoids derived from human pluripotent stem cells (hPSCs). Mature kidney organoids contain nephrons and stromal cells with fibrogenic potential, spatially organized in a manner that resembles the anatomy of the kidney. Organoid nephron damage and interstitial fibrosis can be induced under well-controlled experimental conditions in vitro, making this an ideal system for the study of tissue-intrinsic cell signaling and intercellular crosstalk mechanisms in the absence of systemic signals and immune cells that are present in vivo. Here we describe methods for the generation of kidney organoids from a widely used hPSC line, and for the induction and analysis of nephron damage and interstitial fibrosis.


Subject(s)
Cell Culture Techniques/methods , Kidney/pathology , Organoids/pathology , Pluripotent Stem Cells/cytology , Cell Communication , Cell Line , Fibrosis , Genetic Markers , Humans , Kidney/chemistry , Microscopy, Fluorescence , Organoids/chemistry , Organoids/cytology , Pluripotent Stem Cells/chemistry , Pluripotent Stem Cells/pathology , Real-Time Polymerase Chain Reaction , Signal Transduction
12.
Sci Rep ; 10(1): 18921, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33144601

ABSTRACT

The role of miRNAs in intestinal lipid metabolism is poorly described. The small intestine is constantly exposed to high amounts of dietary lipids, and it is under conditions of stress that the functions of miRNAs become especially pronounced. Approaches consisting in either a chronic exposure to cholesterol and triglyceride rich diets (for several days or weeks) or an acute lipid challenge were employed in the search for intestinal miRNAs with a potential role in lipid metabolism regulation. According to our results, changes in miRNA expression in response to fat ingestion are dependent on factors such as time upon exposure, gender and small intestine section. Classic and recent intestinal in vitro models (i.e. differentiated Caco-2 cells and murine organoids) partially mirror miRNA modulation in response to lipid challenges in vivo. Moreover, intestinal miRNAs might play a role in triglyceride absorption and produce changes in lipid accumulation in intestinal tissues as seen in a generated intestinal Dicer1-deletion murine model. Overall, despite some variability between the different experimental cohorts and in vitro models, results show that some miRNAs analysed here are modulated in response to dietary lipids, hence likely to participate in the regulation of lipid metabolism, and call for further research.


Subject(s)
Dietary Fats/pharmacology , Intestines/drug effects , MicroRNAs/genetics , Organoids/drug effects , Adult Stem Cells/chemistry , Adult Stem Cells/cytology , Adult Stem Cells/drug effects , Animals , Caco-2 Cells , Cell Differentiation/drug effects , Cells, Cultured , DEAD-box RNA Helicases/genetics , Female , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Intestines/chemistry , Intestines/cytology , Lipid Metabolism , Male , Mice , Organoids/chemistry , Organoids/cytology , Ribonuclease III/genetics , Sequence Analysis, RNA , Sex Characteristics , Time Factors
13.
Sci Rep ; 10(1): 16018, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32994409

ABSTRACT

2D and 3D cultures of 3T3-L1 cells were employed in a study of the effects of Omidenepag (OMD), interacting with a non-prostanoid EP2 receptor, on adipogenesis. Upon adipogenesis, the effects on lipid staining, the mRNA expression of adipogenesis-related genes (Pparγ, CEBPa, Ap2, and Glut4) and the extracellular matrix (ECM) including collagen type 1, 4 and 6, and fibronectin, and the size and physical property of 3D organoids were compared between groups that had been treated with EP2 agonists (butaprost and OMD) and PGF2α. Upon adipogenesis, these significantly suppressed lipid staining and the mRNA expression of related genes. EP2 agonists and PGF2α influenced the mRNA expression of ECM in different manners, and these effects were also different between 2 and 3D cultures. Examining the physical properties by a microsqueezer indicated that the solidity of the 3D organoids became significantly lowered upon adipogenesis and these effects were not affected by EP2 agonists. In contrast, 3D organoid stiffness was markedly enhanced by the presence of PGF2α. These observations indicate that EP2 agonists affect the adipogenesis of 3T3-L1 cells in different manners, as compared to PGF2α, suggesting that OMD may not induce PGF2α related orbital fat atrophy, called the deepening of the upper eyelid sulcus (DUES).


Subject(s)
Adipogenesis/drug effects , Dinoprost/pharmacology , Glycine/analogs & derivatives , Organoids/cytology , Pyrazoles/pharmacology , Pyridines/pharmacology , 3T3-L1 Cells , Animals , Cell Culture Techniques , Extracellular Matrix/drug effects , Extracellular Matrix/genetics , Gene Expression Regulation/drug effects , Gene Regulatory Networks/drug effects , Glycine/pharmacology , Mice , Organ Size/drug effects , Organoids/chemistry , Organoids/drug effects
14.
BMC Microbiol ; 20(1): 205, 2020 07 11.
Article in English | MEDLINE | ID: mdl-32652929

ABSTRACT

BACKGROUND: The intestinal tract undergoes a period of cellular maturation during early life, primarily characterized by the organization of epithelial cells into specialized crypt and villus structures. These processes are in part mediated by the acquisition of microbes. Infants delivered at term typically harbor a stable, low diversity microbiota characterized by an overrepresentation of various Bacilli spp., while pre-term infants are colonized by an assortment of bacteria during the first several weeks after delivery. However, the functional effects of these changes on intestinal epithelium homeostasis and maturation remain unclear. To study these effects, human neonate feces were obtained from term and pre-term infants. Fecal 16S rDNA sequencing and global untargeted LC-MS were performed to characterize microbial composition and metabolites from each population. Murine enteral organoids (enteroids) were cultured with 0.22 µm filtered stool supernatant pooled from term or pre-term infants. RESULTS: Term and pre-term microbial communities differed significantly from each other by principle components analysis (PCoA, PERMANOVA p < 0.001), with the pre-term microbiome characterized by increased OTU diversity (Wilcox test p < 0.01). Term communities were less diverse and dominated by Bacilli (81.54%). Pre-term stools had an increased abundance of vitamins, amino acid derivatives and unconjugated bile acids. Pathway analysis revealed a significant increase in multiple metabolic pathways in pre-term samples mapped to E. coli using the KEGG database related to the fermentation of various amino acids and vitamin biosynthesis. Enteroids cultured with supernatant from pre-term stools proliferated at a higher rate than those cultured with supernatant from term stools (cell viability: 207% vs. 147.7%, p < 0.01), grew larger (area: 81,189µm2 vs. 41,777µm2, p < 0.001), and bud at a higher rate (6.5 vs. 4, p < 0.01). Additionally, genes involved in stem cell proliferation were upregulated in pre-term stool treated enteroid cultures (Lgr5, Ephb2, Ascl2 Sox9) but not term stool treated enteroids. CONCLUSIONS: Our findings indicate that microbial metabolites from the more diverse gut microbiome associated with pre-term infants facilitate stem cell proliferation. Therefore, perturbations of the pre-term microbiota may impair intestinal homeostasis.


Subject(s)
Bacteria/classification , Enterocytes/cytology , Metabolomics/methods , Premature Birth/microbiology , RNA, Ribosomal, 16S/genetics , Animals , Animals, Newborn , Bacteria/chemistry , Bacteria/genetics , Bacteria/isolation & purification , Biomarkers/metabolism , Cell Proliferation , DNA, Bacterial/genetics , DNA, Ribosomal/genetics , Enterocytes/microbiology , Feces/microbiology , Gastrointestinal Microbiome , Gene Expression Regulation , Humans , Infant, Newborn , Mice , Organ Culture Techniques , Organoids/chemistry , Organoids/cytology , Organoids/microbiology , Phylogeny , Term Birth
15.
EBioMedicine ; 56: 102786, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32460166

ABSTRACT

BACKGROUND: Pancreatic patient-derived organoids (PDOs) are a well-established model for studying pancreatic ductal adenocarcinoma (PDAC) carcinogenesis and are potential predictors of clinical responses to chemotherapy. Oncolytic virotherapy is envisioned as a novel treatment modality for pancreatic cancer, and candidate viruses are being tested in clinical trials. Here, we explore the feasibility of using PDOs as a screening platform for the oncolytic adenovirus (OA) response. METHODS: Organoids were established from healthy pancreas and PDAC tissues and assessed for infectivity, oncoselectivity, and patient-dependent sensitivity to OA. Antitumour effects were studied in vivo in organoid xenografts. Further evaluation of oncolytic responses was conducted in organoids derived from orthotopic models or metastastic tissues. FINDINGS: Oncolytic adenoviruses display good selectivity, with replication only in organoids derived from PDAC tumours. Furthermore, responses of PDOs to a set of OAs reveal individual differences in cytotoxicity as well as in synergism with standard chemotherapy. Adenoviral cytotoxicity in PDOs is predictive of antitumour efficacy in a subcutaneous xenograft setting. Organoids from orthotopic tumours and metastases in nude mice mirror the viral preference of PDOs, indicating that PDO sensitivity to OAs could be informative about responses in both primary tumours and metastatic foci. INTERPRETATION: Our data imply that pancreatic PDOs can serve as predictive tools for screening for sensitivity to OA.


Subject(s)
Carcinoma, Pancreatic Ductal/therapy , Oncolytic Viruses/physiology , Organoids/virology , Pancreatic Neoplasms/therapy , Adenoviridae/physiology , Animals , Apoptosis , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation , Drug Screening Assays, Antitumor , Feasibility Studies , Humans , Mice , Mice, Nude , Mutation , Neoplasm Metastasis , Oncolytic Virotherapy , Organoids/chemistry , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Virus Internalization , Xenograft Model Antitumor Assays
16.
Nat Commun ; 11(1): 1711, 2020 04 06.
Article in English | MEDLINE | ID: mdl-32249764

ABSTRACT

Recently, organoid technology has been used to generate a large repository of breast cancer organoids. Here we present an extensive evaluation of the ability of organoid culture technology to preserve complex stem/progenitor and differentiated cell types via long-term propagation of normal human mammary tissues. Basal/stem and luminal progenitor cells can differentiate in culture to generate mature basal and luminal cell types, including ER+ cells that have been challenging to maintain in culture. Cells associated with increased cancer risk can also be propagated. Single-cell analyses of matched organoid cultures and native tissues by mass cytometry for 38 markers provide a higher resolution representation of the multiple mammary epithelial cell types in the organoids, and demonstrate that protein expression patterns of the tissue of origin can be preserved in culture. These studies indicate that organoid cultures provide a valuable platform for studies of mammary differentiation, transformation, and breast cancer risk.


Subject(s)
Cell Culture Techniques/methods , Cell Lineage , Mammary Glands, Human/cytology , Organoids/cytology , Organoids/metabolism , Stem Cells/cytology , Adult , BRCA1 Protein/genetics , Breast Neoplasms , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Lineage/genetics , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Female , Humans , Mammary Glands, Human/chemistry , Mammary Glands, Human/metabolism , Middle Aged , Organoids/chemistry , Single-Cell Analysis , Stem Cells/chemistry , Stem Cells/metabolism , Transforming Growth Factor beta/antagonists & inhibitors , Young Adult , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
17.
J Ocul Pharmacol Ther ; 36(1): 25-29, 2020.
Article in English | MEDLINE | ID: mdl-31166829

ABSTRACT

In this study, we describe efforts by the National Eye Institute (NEI) and National Center for Advancing Translational Science (NCATS) to catalyze advances in 3-dimensional (3-D) ocular organoid and microphysiological systems (MPS). We reviewed the recent literature regarding ocular organoids and tissue chips. Animal models, 2-dimensional cell culture models, and postmortem human tissue samples provide the vision research community with insights critical to understanding pathophysiology and therapeutic development. The advent of induced pluripotent stem cell technologies provide researchers with enticing new approaches and tools that augment study in more traditional models to provide the scientific community with insights that have previously been impossible to obtain. Efforts by the National Institutes of Health (NIH) have already accelerated the pace of scientific discovery, and recent advances in ocular organoid and MPS modeling approaches have opened new avenues of investigation. In addition to more closely recapitulating the morphologies and physiological responses of in vivo human tissue, key breakthroughs have been made in the past year to resolve long-standing scientific questions regarding tissue development, molecular signaling, and pathophysiological mechanisms that promise to provide advances critical to therapeutic development and patient care. 3-D tissue culture modeling and MPS offer platforms for future high-throughput testing of therapeutic candidates and studies of gene interactions to improve models of complex genetic diseases with no well-defined etiology, such as age-related macular degeneration and Fuchs' dystrophy.


Subject(s)
Drug Development , Induced Pluripotent Stem Cells/metabolism , Lab-On-A-Chip Devices , Models, Biological , Ophthalmic Solutions/chemical synthesis , Organoids/metabolism , Animals , Humans , Induced Pluripotent Stem Cells/chemistry , Ophthalmic Solutions/chemistry , Organoids/chemistry , Tissue Engineering
18.
Colloids Surf B Biointerfaces ; 186: 110708, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31838268

ABSTRACT

Emulsion-based generation of hydrogel particles has been widely explored for numerous applications in fields such as biomedical, food, and drug delivery. Water-in-water emulsion (w/w) is an organic solvent-free approach and exploits solely aqueous media to generate nano- or microparticles. This strategy is environment-friendly and favorable for biomedical applications where biocompatibility is the ultimate criterion. Hence, PEG-based microgels can be synthesized with desired size and functionality using w/w emulsion technique. To estimate the influence of emulsification parameters on size and stability of PEG-lipid microgels, optimizations using three independent input variables were carried out: (i) ultrasonication power, (ii) ultrasonication duration, and (iii) duration of light exposure. Physical immobilization of microgels on islet-organoids was achieved through hydrophobic interactions. Cell function and viability were assessed thoroughly after microgel immobilization. Microgel size is dependent on ultrasonication parameters and microgel stability is vastly determined by the duration of light exposure. Immobilization of microgels with 5 mM lipid moiety promoted coating of islet-organoids. Coated organoids retained their function and viability without significant adverse effects. This is important for understanding fundamental aspects of PEG-lipid microgels using w/w emulsion, useful for possible drug/gene delivery applications to increase treatment efficiency and ultimately lead to clinical translation of PEG microgels for biomedical applications.


Subject(s)
Lipids/chemistry , Microgels/chemistry , Organoids/chemistry , Polyethylene Glycols/chemistry , Particle Size , Surface Properties , Water/chemistry
19.
J Vis Exp ; (152)2019 10 13.
Article in English | MEDLINE | ID: mdl-31657789

ABSTRACT

Dietary lipids are taken up as free fatty acids (FAs) by the intestinal epithelium. These FAs are intracellularly converted into triglyceride (TG) molecules, before they are packaged into chylomicrons for transport to the lymph or into cytosolic lipid droplets (LDs) for intracellular storage. A crucial step for the formation of LDs is the catalytic activity of diacylglycerol acyltransferases (DGAT) in the final step of TG synthesis. LDs are important to buffer toxic lipid species and regulate cellular metabolism in different cell types. Since the human intestinal epithelium is regularly confronted with high concentrations of lipids, LD formation is of great importance to regulate homeostasis. Here we describe a simple assay for the characterization and quantification of LD formation (LDF) upon stimulation with the most common unsaturated fatty acid, oleic acid, in human intestinal organoids. The LDF assay is based on the LD-specific fluorescent dye LD540, which allows for quantification of LDs by confocal microscopy, fluorescent plate reader, or flow cytometry. The LDF assay can be used to characterize LD formation in human intestinal epithelial cells, or to study human (genetic) disorders that affect LD metabolism, such as DGAT1 deficiency. Furthermore, this assay can also be used in a high-throughput pipeline to test novel therapeutic compounds, which restore defects in LD formation in intestinal or other types of organoids.


Subject(s)
Fluorescent Dyes/analysis , Intestinal Mucosa/chemistry , Intestine, Small/chemistry , Lipid Droplets/chemistry , Lipid Metabolism/physiology , Organoids/chemistry , Cells, Cultured , Fluorescence , Humans , Intestinal Mucosa/metabolism , Intestine, Small/cytology , Lipid Droplets/metabolism , Organoids/metabolism
20.
Biofabrication ; 12(1): 015002, 2019 10 21.
Article in English | MEDLINE | ID: mdl-31487700

ABSTRACT

The high attrition rate of neuro-pharmaceuticals as they proceed to market necessitates the development of clinically-relevant in vitro neural microphysiological systems that can be utilized during the preclinical screening phase to assess the safety and efficacy of potential compounds. Historically, proposed models have adhered to two distinct approaches; those that are biologically relevant (e.g.-organoids, spheroids) or those that provide engineering control (e.g.-bioprinting, microfluidics). Separately, these approaches fail to fully recapitulate the complex hierarchical structure of the nervous system, limiting their clinical applications. Furthermore, the reliance on manual implementation present in many models fails to effectively scale up or satisfy the consistency standards required for widespread industry adoption. This work serves as a proof-of-concept for merging the two approaches to create a neural microphysiological system that overcomes their individual limitations. Spinal cord spheroids, fabricated using magnetic nanoparticles, are positioned in a three-dimensional hydrogel construct using magnetic bioprinting. Resulting constructs demonstrate both localized cell-cell interactions and long-distance projections that mimic in vivo structure. The use of magnetic nanoparticles for spheroid formation provides batch-to-batch consistency in size and shape and reduces the reliance on trained experimenters for accurate placing for culture. Taken together, this combination approach provides the first steps towards developing a simple approach for integrating spheroid, hydrogel culture, and bioprinting as an alternative to more specialized and expensive processes.


Subject(s)
Bioprinting/methods , Spinal Cord/cytology , Bioprinting/instrumentation , Cell Proliferation , Humans , Hydrogels/chemistry , Magnetics , Nanoparticles/chemistry , Organoids/chemistry , Spheroids, Cellular/chemistry , Spheroids, Cellular/cytology , Spinal Cord/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...