Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.756
Filter
1.
Biomaterials ; 312: 122719, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39088912

ABSTRACT

Acute myeloid leukemia (AML) is a deadly form of leukemia with ineffective traditional treatment and frequent chemoresistance-associated relapse. Personalized drug screening holds promise in identifying optimal regimen, nevertheless, primary AML cells undergo spontaneous apoptosis during cultures, invalidating the drug screening results. Here, we reconstitute a 3D osteogenic niche (3DON) mimicking that in bone marrow to support primary AML cell survival and phenotype maintenance in cultures. Specifically, 3DON derived from osteogenically differentiated mesenchymal stem cells (MSC) from healthy and AML donors are co-cultured with primary AML cells. The AML cells under the AML_3DON niche showed enhanced viability, reduced apoptosis and maintained CD33+ CD34-phenotype, associating with elevated secretion of anti-apoptotic cytokines in the AML_3DON niche. Moreover, AML cells under the AML_3DON niche exhibited low sensitivity to two FDA-approved chemotherapeutic drugs, further suggesting the physiological resemblance of the AML_3DON niche. Most interestingly, AML cells co-cultured with the healthy_3DON niche are highly sensitive to the same sample drugs. This study demonstrates the differential responses of AML cells towards leukemic and healthy bone marrow niches, suggesting the impact of native cancer cell niche in drug screening, and the potential of re-engineering healthy bone marrow niche in AML patients as chemotherapeutic adjuvants overcoming chemoresistance, respectively.


Subject(s)
Cell Survival , Leukemia, Myeloid, Acute , Mesenchymal Stem Cells , Phenotype , Tumor Microenvironment , Humans , Leukemia, Myeloid, Acute/pathology , Tumor Microenvironment/drug effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Cell Survival/drug effects , Coculture Techniques/methods , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bone Marrow/pathology , Bone Marrow/drug effects , Stem Cell Niche/drug effects , Bone Marrow Cells/cytology , Male , Cell Differentiation/drug effects , Female
2.
PLoS One ; 19(9): e0310314, 2024.
Article in English | MEDLINE | ID: mdl-39312543

ABSTRACT

In vitro co-culture techniques that allow the growth of plants and pathogens under controlled environmental conditions are being used to re-create host plant infection. These approaches reduce infection times, promote reproducibility, and enable a rapid evaluation of plant-pathogen interactions. As a result, these systems have become essential in breeding programs aimed at developing plant resistance to diseases. In this study, we developed and validated an in vitro co-culture system to investigate the Armillaria root rot (ARR) affecting Prunus spp. This disease, caused by fungi Armillaria spp. and Desarmillaria caespitosa, poses a severe threat to the stone and nut fruit industry due to the susceptibility of most commercial rootstocks to infection and the lack of effective management options for its control. The system consists of a fiber-supported liquid approach in sterile plastic vessels that allows a fast and reproducible fungal infection under controlled environmental conditions. The floor of the vessels was covered with a polyester-fiber matte and a germination paper that served as an interface between the mycelia and the plant roots. The vessels were subjected to inoculation with Armillaria mellea and D. caespitosa, and three Prunus genotypes ('Guardian®', 'MP-29', and Prunus cerasifera '14-4') were co-cultured with both fungi. Disease progression and plant and fungal biomass were monitored during co-culture. The presented in vitro co-culture approach facilitates the concurrent growth of Armillaria/Desarmillaria spp. and Prunus spp., excluding most of the limitations associated with greenhouses and field experiments. This system provides consistent and reproducible conditions for investigating a prominent plant disease affecting Prunus spp.


Subject(s)
Armillaria , Coculture Techniques , Plant Diseases , Plant Roots , Prunus , Coculture Techniques/methods , Prunus/microbiology , Plant Roots/microbiology , Plant Roots/growth & development , Plant Diseases/microbiology , Armillaria/genetics
3.
Int J Mol Sci ; 25(18)2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39337326

ABSTRACT

Helicobacter pylori is one of the most common bacterial pathogens worldwide and the main etiological agent of numerous gastric diseases. The frequency of multidrug resistance of H. pylori is growing and the leading factor related to this phenomenon is its ability to form biofilm. Therefore, the establishment of a proper model to study this structure is of critical need. In response to this, the aim of this original article is to validate conditions of the optimal biofilm development of H. pylori in monoculture and co-culture with a gastric cell line in media simulating human fluids. Using a set of culture-based and microscopic techniques, we proved that simulated transcellular fluid and simulated gastric fluid, when applied in appropriate concentrations, stimulate autoaggregation and biofilm formation of H. pylori. Additionally, using a co-culture system on semi-permeable membranes in media imitating the stomach environment, we were able to obtain a monolayer of a gastric cell line with H. pylori biofilm on its surface. We believe that the current model for H. pylori biofilm formation in monoculture and co-culture with gastric cells in media containing host-mimicking fluids will constitute a platform for the intensification of research on H. pylori biofilms in in vitro conditions that simulate the human body.


Subject(s)
Biofilms , Coculture Techniques , Helicobacter pylori , Biofilms/growth & development , Biofilms/drug effects , Helicobacter pylori/physiology , Humans , Coculture Techniques/methods , Stomach/microbiology , Helicobacter Infections/microbiology , Cell Line
4.
Mol Biol Rep ; 51(1): 1024, 2024 Sep 28.
Article in English | MEDLINE | ID: mdl-39340590

ABSTRACT

Neurodegenerative diseases (NDs) are characterized by the progressive loss of neurons. As to developing effective therapeutic interventions, it is crucial to understand the underlying mechanisms of NDs. Cellular models have become invaluable tools for studying the complex pathogenesis of NDs, offering insights into disease mechanisms, determining potential therapeutic targets, and aiding in drug discovery. This review provides a comprehensive overview of various cellular models used in ND research, focusing on Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. Cell lines, such as SH-SY5Y and PC12 cells, have emerged as valuable tools due to their ease of use, reproducibility, and scalability. Additionally, co-culture models, involving the growth of distinct cell types like neurons and astrocytes together, are highlighted for simulating brain interactions and microenvironment. While cell lines cannot fully replicate the complexity of the human brain, they provide a scalable method for examining important aspects of neurodegenerative diseases. Advancements in cell line technologies, including the incorporation of patient-specific genetic variants and improved co-culture models, hold promise for enhancing our understanding and expediting the development of effective treatments. Integrating multiple cellular models and advanced technologies offers the potential for significant progress in unraveling the intricacies of these debilitating diseases and improving patient outcomes.


Subject(s)
Neurodegenerative Diseases , Neurons , Humans , Neurodegenerative Diseases/pathology , Neurons/metabolism , Animals , Coculture Techniques/methods , Cell Line , Models, Biological , Alzheimer Disease/pathology , Alzheimer Disease/genetics
5.
An Acad Bras Cienc ; 96(suppl 1): e20230633, 2024.
Article in English | MEDLINE | ID: mdl-39319832

ABSTRACT

Biorefineries require low-cost production processes, low waste generation and equipment that can be used not only for a single process, but for the manufacture of several products. In this context, in this research a continuous 3D printing microbioreactor coupled to an Arduino-controlled automatic feeding system was developed for the intensification of the ethanol production process from xylose/xylulose (3:1), using a new biocatalyst containing the co-culture of Scheffersomyces stipitis and Saccharomyces cerevisiae (50/50). Initially, batch fermentations of monocultures of S. cerevisiae and S. stipitis and co-culture were carried out. Subsequently, the immobilized co-culture was used as a biocatalyst in continuous fermentations using the developed microreactor. Fermentations carried out in the microbioreactor presented a 2-fold increase in the ethanol concentration and a 3-fold increase in productivity when compared to monocultures. The microbioreactor developed proved to be efficient and can be extended for other bioproducts production. This approach proved to be a promising alternative for the use of the hemicellulose fraction of biomasses without the need to use modified strains.


Subject(s)
Bioreactors , Coculture Techniques , Ethanol , Fermentation , Printing, Three-Dimensional , Saccharomyces cerevisiae , Saccharomycetales , Ethanol/metabolism , Saccharomyces cerevisiae/metabolism , Coculture Techniques/methods , Bioreactors/microbiology , Saccharomycetales/metabolism , Saccharomycetales/growth & development
6.
Curr Protoc ; 4(9): e70013, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39269316

ABSTRACT

The study of human intestinal physiology and host-microbe interactions is crucial for understanding gastrointestinal health and disease. Traditional two-dimensional cell culture models lack the complexity of the native intestinal environment, limiting their utility in studying intestinal biology. Here, we present a detailed protocol for the set up and utilization of a three-dimensional (3D) in vitro bioreactor system for human intestinal studies and bacterial co-culture. This article outlines the design and assembly of the bioreactor system, scaffold fabrication, bacterial culture techniques, analysis methods, and troubleshooting tips. By providing step-by-step instructions, the goal is to enable other laboratories to utilize physiologically relevant tissue models of the human intestine, incorporating key features, such as nutrient flow, multiple human cell types, 3D architecture, and microbial communities. The incorporation of commensal bacteria into the bioreactor system allows for the investigation of complex host-microbe interactions, providing insight into gastrointestinal health and pathology. This article serves as a comprehensive resource for scientists seeking to advance their understanding of intestinal biology toward the development of novel therapeutic strategies for gastrointestinal disorders. © 2024 Wiley Periodicals LLC. Basic Protocol 1: Scaffold design Basic Protocol 2: Intestinal cell culture: Caco2 cells Basic Protocol 3: Intestinal cell culture: organoids Basic Protocol 4: Bioreactor design and set up Basic Protocol 5: Bacteria in 3D bioreactor set up Basic Protocol 6: Bacteria and drug dosing.


Subject(s)
Bioreactors , Coculture Techniques , Intestines , Humans , Bioreactors/microbiology , Coculture Techniques/methods , Coculture Techniques/instrumentation , Intestines/microbiology , Intestines/cytology , Caco-2 Cells , Gastrointestinal Microbiome , Cell Culture Techniques, Three Dimensional/methods , Cell Culture Techniques, Three Dimensional/instrumentation
7.
Stem Cell Res Ther ; 15(1): 273, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39218985

ABSTRACT

BACKGROUND: Chronic lung disease of prematurity, called bronchopulmonary dysplasia (BPD), lacks effective therapies, stressing the need for preclinical testing systems that reflect human pathology for identifying causal pathways and testing novel compounds. Alveolar organoids derived from human pluripotent stem cells (hPSC) are promising test platforms for studying distal airway diseases like BPD, but current protocols do not accurately replicate the distal niche environment of the native lung. Herein, we investigated the contributions of cellular constituents of the alveolus and fetal respiratory movements on hPSC-derived alveolar organoid formation. METHODS: Human PSCs were differentiated in 2D culture into lung progenitor cells (LPC) which were then further differentiated into alveolar organoids before and after removal of co-developing mesodermal cells. LPCs were also differentiated in Transwell® co-cultures with and without human fetal lung fibroblast. Forming organoids were subjected to phasic mechanical strain using a Flexcell® system. Differentiation within organoids and Transwell® cultures was assessed by flow cytometry, immunofluorescence, and qPCR for lung epithelial and alveolar markers of differentiation including GATA binding protein 6 (GATA 6), E-cadherin (CDH1), NK2 Homeobox 1 (NKX2-1), HT2-280, surfactant proteins B (SFTPB) and C (SFTPC). RESULTS: We observed that co-developing mesenchymal progenitors promote alveolar epithelial type 2 cell (AEC2) differentiation within hPSC-derived lung organoids. This mesenchymal effect on AEC2 differentiation was corroborated by co-culturing hPSC-NKX2-1+ lung progenitors with human embryonic lung fibroblasts. The stimulatory effect did not require direct contact between fibroblasts and NKX2-1+ lung progenitors. Additionally, we demonstrate that episodic mechanical deformation of hPSC-derived lung organoids, mimicking in situ fetal respiratory movements, increased AEC2 differentiation without affecting proximal epithelial differentiation. CONCLUSION: Our data suggest that biophysical and mesenchymal components promote AEC2 differentiation within hPSC-derived distal organoids in vitro.


Subject(s)
Cell Differentiation , Lung , Organoids , Humans , Organoids/cytology , Organoids/metabolism , Lung/cytology , Lung/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Coculture Techniques/methods , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism
8.
Sci Rep ; 14(1): 20703, 2024 09 05.
Article in English | MEDLINE | ID: mdl-39237637

ABSTRACT

This work uses response surface methodology (RSM) to study the co-cultivation of symbiotic indigenous wastewater microalgae and bacteria under different conditions (inoculum ratio of bacteria to microalgae, CO2, light intensity, and harvest time) for optimal bioenergy feedstock production. The findings of this study demonstrate that the symbiotic microalgae-bacteria culture not only increases total microalgal biomass and lipid productivity, but also enlarges microalgal cell size and stimulates lipid accumulation. Meanwhile, inoculum ratio of bacteria to microalgae, light intensity, CO2, and harvest time significantly affect biomass and lipid productivity. CO2 concentration and harvest time have significant interactive effect on lipid productivity. The response of microalgal biomass and lipid productivity varies significantly from 2.1 × 105 to 1.9 × 107 cells/mL and 2.8 × 102 to 3.7 × 1012 Total Fluorescent Units/mL respectively. Conditions for optimum biomass and oil accumulation are 100% of inoculation ratio (bacteria/microalgae), 3.6% of CO2 (v/v), 205.8 µmol/m2/s of light intensity, and 10.6 days of harvest time. This work provides a systematic methodology with RSM to explore the benefits of symbiotic microalgae-bacteria culture, and to optimize various cultivation parameters within complex wastewater environments for practical applications of integrated wastewater-microalgae systems for cost-efficient bioenergy production.


Subject(s)
Bacteria , Biofuels , Biomass , Carbon Dioxide , Microalgae , Wastewater , Wastewater/microbiology , Microalgae/growth & development , Microalgae/metabolism , Biofuels/microbiology , Bacteria/metabolism , Bacteria/growth & development , Carbon Dioxide/metabolism , Coculture Techniques/methods , Symbiosis , Lipids/biosynthesis , Lipids/analysis
9.
J Biomed Sci ; 31(1): 84, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39180048

ABSTRACT

BACKGROUND: Identification of lung cancer subtypes is critical for successful treatment in patients, especially those in advanced stages. Many advanced and personal treatments require knowledge of specific mutations, as well as up- and down-regulations of genes, for effective targeting of the cancer cells. While many studies focus on individual cell structures and delve deeper into gene sequencing, the present study proposes a machine learning method for lung cancer classification based on low-magnification cancer outgrowth patterns in a 2D co-culture environment. METHODS: Using a magnetic well plate holder, circular pattern lung cancer cell clusters were generated among fibroblasts, and daily images were captured to monitor cancer outgrowth over a 9-day period. These outgrowth images were then augmented and used to train a convolutional neural network (CNN) model based on the lightweight TinyVGG architecture. The model was trained with pairs of classes representing three subtypes of NSCLC: A549 (adenocarcinoma), H520 (squamous cell carcinoma), and H460 (large cell carcinoma). The objective was to assess whether this lightweight machine learning model could accurately classify the three lung cancer cell lines at different stages of cancer outgrowth. Additionally, cancer outgrowth images of two patient-derived lung cancer cells, one with the KRAS oncogene and the other with the EGFR oncogene, were captured and classified using the CNN model. This demonstration aimed to investigate the translational potential of machine learning-enabled lung cancer classification. RESULTS: The lightweight CNN model achieved over 93% classification accuracy at 1 day of outgrowth among A549, H460, and H520, and reached 100% classification accuracy at 7 days of outgrowth. Additionally, the model achieved 100% classification accuracy at 4 days for patient-derived lung cancer cells. Although these cells are classified as Adenocarcinoma, their outgrowth patterns vary depending on their oncogene expressions (KRAS or EGFR). CONCLUSIONS: These results demonstrate that the lightweight CNN architecture, operating locally on a laptop without network or cloud connectivity, can effectively create a machine learning-enabled model capable of accurately classifying lung cancer cell subtypes, including those derived from patients, based upon their outgrowth patterns in the presence of surrounding fibroblasts. This advancement underscores the potential of machine learning to enhance early lung cancer subtyping, offering promising avenues for improving treatment outcomes in advanced stage-patients.


Subject(s)
Coculture Techniques , Fibroblasts , Lung Neoplasms , Machine Learning , Neural Networks, Computer , Humans , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Cell Line, Tumor , Coculture Techniques/methods , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology
10.
Xenotransplantation ; 31(4): e12878, 2024.
Article in English | MEDLINE | ID: mdl-39166823

ABSTRACT

Hepatocyte transplantation and bioartificial liver (BAL) systems hold significant promise as less invasive alternatives to traditional transplantation, providing crucial temporary support for patients with acute and chronic liver failure. Although human hepatocytes are ideal, their use is limited by ethical concerns and donor availability, leading to the use of porcine hepatocytes in BAL systems due to their functional similarities. Recent advancements in gene-editing technology have improved porcine organ xenotransplantation clinical trials by addressing immune rejection issues. Gene-edited pigs, such as alpha-1,3-galactosyltransferase (GGTA1) knockout pigs, offer a secure source of primary cells for BAL systems. Our research focuses on optimizing the safety and functionality of porcine primary hepatocytes during large-scale cultivation. We achieved this by creating GGTA1 knockout pigs through one-step delivery of CRISPR/Cas9 to pig zygotes via oviduct injection of rAAV, and enhancing hepatocyte viability and function by co-culturing hepatocytes with Roof plate-specific spondin 1 overexpressing HUVECs (R-HUVECs). Using a Rocker culture system, approximately 1010 primary porcine hepatocytes and R-HUVECs rapidly formed organoids with a diameter of 92.1 ± 28.1 µm within 24 h. These organoids not only maintained excellent functionality but also supported partial hepatocyte self-renewal during long-term culture over 28 days. Gene-edited primary porcine hepatocyte organoids will significantly advance the applications of hepatocyte transplantation and BAL systems.


Subject(s)
Galactosyltransferases , Gene Editing , Hepatocytes , Liver, Artificial , Organoids , Transplantation, Heterologous , Animals , Galactosyltransferases/genetics , Swine , Transplantation, Heterologous/methods , Organoids/metabolism , Gene Editing/methods , Humans , Animals, Genetically Modified , CRISPR-Cas Systems , Gene Knockout Techniques/methods , Coculture Techniques/methods
11.
Biotechnol J ; 19(8): e2400070, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39167558

ABSTRACT

Organoids have emerged as valuable tools for the study of development and disease. Assembloids are formed by integrating multiple organoid types to create more complex models. However, the process by which organoids integrate to form assembloids remains unclear and may play an important role in the resulting organoid structure. Here, a microfluidic platform is developed that allows separate culture of distinct organoid types and provides the capacity to partially control the geometry of the resulting organoid surfaces. Removal of a microfabricated barrier then allows the shaped and positioned organoids to interact and form an assembloid. When midbrain and unguided brain organoids were allowed to assemble with a defined spacing between them, axonal projections from midbrain organoids and cell migration out of unguided organoids were observed and quantitatively measured as the two types of organoids fused together. Axonal projection directions were statistically biased toward other midbrain organoids, and unguided organoid surface geometry was found to affect cell invasion. This platform provides a tool to observe cellular interactions between organoid surfaces that are spaced apart in a controlled manner, and may ultimately have value in exploring neuronal migration, axon targeting, and assembloid formation mechanisms.


Subject(s)
Cell Movement , Coculture Techniques , Organoids , Organoids/cytology , Organoids/metabolism , Coculture Techniques/methods , Animals , Cell Movement/physiology , Brain/cytology , Mesencephalon/cytology , Mice , Lab-On-A-Chip Devices , Axons , Microtechnology/methods , Humans , Neurons/cytology
12.
Sci Rep ; 14(1): 19578, 2024 08 23.
Article in English | MEDLINE | ID: mdl-39179636

ABSTRACT

Large-scale production of cultured meat requires bulk culture medium containing growth-promoting proteins from animal serum. However, animal serum for mammalian cell culture is associated with high costs, ethical concerns, and contamination risks. Owing to its growth factor content, conditioned medium from rat liver epithelial RL34 cells can replace animal serum for myoblast proliferation. More seeded cells and longer culture periods are thought to yield higher growth factor levels, resulting in more effective muscle cell proliferation. However, RL34 cells can deplete nutrients and release harmful metabolites into the culture medium over time, potentially causing growth inhibition and apoptosis. This issue highlights the need for waste clearance during condition medium production. To address this issue, we introduced a lactate permease gene (lldP) and an L-lactate-to-pyruvate conversion enzyme gene (lldD) to generate a recombinant L-lactate-assimilating cyanobacterium Synechococcus sp. KC0110 strain. Transwell co-culture of this strain with RL34 cells exhibited a marked reduction in the levels of harmful metabolites, lactate and ammonium, while maintaining higher concentrations of glucose, pyruvate, and pyruvate-derived amino acids than those seen with RL34 cell monocultures. The co-culture medium supported myoblast proliferation without medium dilution or additional nutrients, which was attributed to the waste clearance and nutrient replenishment effects of the KC0110 strain. This culture system holds potential for the production of low-cost, and animal-free cultured meat.


Subject(s)
Coculture Techniques , Lactic Acid , Meat , Animals , Lactic Acid/metabolism , Rats , Coculture Techniques/methods , Culture Media, Serum-Free , Cell Proliferation , Synechococcus/metabolism , Synechococcus/genetics , Synechococcus/growth & development , Cell Line , Myoblasts/metabolism , Myoblasts/cytology , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , In Vitro Meat
13.
Mol Pharm ; 21(9): 4664-4672, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39133897

ABSTRACT

The blood-brain barrier (BBB) poses a significant challenge for drug delivery and is linked to various neurovascular disorders. In vitro BBB models provide a tool to investigate drug permeation across the BBB and the barrier's response to external injury events. Yet, existing models lack fidelity in replicating the BBB's complexity, hindering a comprehensive understanding of its functions. This study introduces a three-dimensional (3D) model using polyethylene glycol (PEG) hydrogels modified with biomimetic peptides that represent recognition sequences of key proteins in the brain. Hydrogels were functionalized with recognition sequences for laminin (IKVAV) and fibronectin peptides (RGD) and chemically cross-linked with matrix metalloprotease-sensitive peptides (MMPs) to mimic the extracellular matrix of the BBB. Astrocytes and endothelial cells were seeded within and on the surface of the hydrogels, respectively. The barrier integrity was assessed through different tests including transendothelial electrical resistance (TEER), the permeability of sodium fluorescence (Na-F), the permeability of Evan's blue bound to albumin (EBA), and the expression of zonula occluden-1 (ZO-1) in seeded endothelial cells. Hydrogels with a combination of RGD and IKVAV peptides displayed superior performance, exhibiting significantly higher TEER values (55.33 ± 1.47 Ω·cm2) at day 5 compared to other 2D controls including HAECs-monoculture and HAECs-cocultured with NHAs seeded on well inserts and 3D controls including RGD hydrogel and RGD-IKVAV monoculture with HAECs and RGD hydrogel cocultured with HAECs and NHAs. The designed 3D system resulted in the lowest Evan's blue permeability at 120 min (0.215 ± 0.055 µg/mL) compared to controls. ZO-1 expression was significantly higher and formed a relatively larger network in the functionalized hydrogel cocultured with astrocytes and endothelial cells compared to the controls. Thus, the designed 3D model effectively recapitulates the main BBB structure and function in vitro and is expected to contribute to a deeper understanding of pathological CNS angiogenesis and the development of effective CNS medications.


Subject(s)
Astrocytes , Blood-Brain Barrier , Coculture Techniques , Endothelial Cells , Hydrogels , Peptides , Polyethylene Glycols , Blood-Brain Barrier/metabolism , Astrocytes/metabolism , Polyethylene Glycols/chemistry , Endothelial Cells/metabolism , Coculture Techniques/methods , Hydrogels/chemistry , Peptides/chemistry , Humans , Oligopeptides/chemistry , Fibronectins/chemistry , Fibronectins/metabolism , Laminin/chemistry , Animals , Biomimetics/methods , Biomimetic Materials/chemistry , Cells, Cultured
14.
Analyst ; 149(18): 4675-4686, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39086194

ABSTRACT

This study presents the development and validation of an innovative microfluidic liver-on-a-chip device utilizing gravity-driven perfusion for the evaluation of drug hepatotoxicity. This research involved the construction of a hydrogel-based coculture chip that integrates liver parenchymal and stellate cells within a tri-channel configuration. The assembly and operation of the liver-on-a-chip and its accompanying custom rocker were straightforward. The cells in the chip maintained high viability and continuously synthesized liver albumin over extended culture durations. Acetaminophen (APAP), a hepatic injury-inducing drug, was utilized as a positive control in hepatic toxicity assays on the chip. The liver chip exhibited hepatotoxic responses comparable to those observed in 2D models. Furthermore, in this study we evaluated the effects of two plant-derived natural compounds, aristolochic acid I (AA) and its analog aristolactam AII (AL), in both 2D cell models and the liver-on-a-chip system. AA, known for its hepatorenal toxicity, was observed to cause hepatotoxicity in both the 2D models and on the chip. The flow cytometry and mRNA sequencing results confirmed the propensity of these compounds to induce liver cell apoptosis. Notably, AL, previously considered nontoxic, provoked a significant decrease in the hepatic functionality marker albumin exclusively in the liver chip but not in 2D models, indicating the liver chip's enhanced sensitivity to toxic substances. In summary, this pumpless liver-on-a-chip is a simple yet powerful tool for drug hepatotoxicity studies.


Subject(s)
Acetaminophen , Lab-On-A-Chip Devices , Liver , Humans , Acetaminophen/toxicity , Liver/drug effects , Liver/pathology , Aristolochic Acids/toxicity , Apoptosis/drug effects , Chemical and Drug Induced Liver Injury/pathology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Cell Survival/drug effects , Hep G2 Cells , Coculture Techniques/methods
15.
Bioresour Technol ; 410: 131232, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39117247

ABSTRACT

Applying low-cost substrate is critical for sustainable bioproduction. Co-culture of phototrophic and heterotrophic microorganisms can be a promising solution as they can use CO2 and light as feedstock. This study aimed to create a light-driven consortium using a marine cyanobacterium Synechococcus sp. PCC 7002 and an industrial yeast Yarrowia lipolytica. First, the cyanobacterium was engineered to accumulate and secrete sucrose by regulating the expression of genes involved in sucrose biosynthesis and transport, resulting in 4.0 g/L of sucrose secretion. Then, Yarrowia lipolytica was engineered to efficiently use sucrose and produce ß-caryophyllene that has various industrial applications. Then, co- and sequential-culture were optimized with different induction conditions and media compositions. A maximum ß-caryophyllene yield of 14.1 mg/L was obtained from the co-culture. This study successfully established an artificial light-driven consortium based on a marine cyanobacterium and Y. lipolytica, and provides a foundation for sustainable bioproduction from CO2 and light through co-culture systems.


Subject(s)
Coculture Techniques , Light , Polycyclic Sesquiterpenes , Synechococcus , Yarrowia , Coculture Techniques/methods , Polycyclic Sesquiterpenes/metabolism , Synechococcus/metabolism , Synechococcus/growth & development , Yarrowia/metabolism , Sucrose/metabolism , Sesquiterpenes/metabolism , Heterotrophic Processes , Autotrophic Processes
16.
Nat Commun ; 15(1): 7424, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39198465

ABSTRACT

In fetal development, tissue interaction such as the interplay between blood vessel (BV) and epithelial tissue is crucial for organogenesis. Here we recapitulate the spatial arrangement between liver epithelial tissue and the portal vein to observe the formation of intrahepatic bile ducts (BDs) from human induced pluripotent stem cells (hiPSC). We co-culture hiPSC-liver progenitors on the artificial BV consisting of immature smooth muscle cells and endothelial cells, both derived from hiPSCs. After 3 weeks, liver progenitors within hiPSC-BV-incorporated liver organoids (BVLO) differentiate to cholangiocytes and acquire epithelial characteristics, including intercellular junctions, microvilli on the apical membrane, and secretory functions. Furthermore, liver surface transplanted-BVLO temporarily attenuates cholestatic injury symptoms. Single cell RNA sequence analysis suggests that BD interact with the BV in BVLO through TGFß and Notch pathways. Knocking out JAG1 in hiPSC-BV significantly attenuates bile duct formation, highlighting BVLO potential as a model for Alagille syndrome, a congenital biliary disease. Overall, we develop a novel 3D co-culture method that successfully establishes functional human BDs by emulating liver epithelial-BV interaction.


Subject(s)
Cell Differentiation , Coculture Techniques , Induced Pluripotent Stem Cells , Jagged-1 Protein , Liver , Organoids , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Organoids/metabolism , Organoids/cytology , Liver/cytology , Liver/metabolism , Liver/blood supply , Coculture Techniques/methods , Jagged-1 Protein/metabolism , Jagged-1 Protein/genetics , Alagille Syndrome/genetics , Alagille Syndrome/metabolism , Animals , Bile Ducts, Intrahepatic/cytology , Bile Ducts, Intrahepatic/metabolism , Blood Vessels/cytology , Blood Vessels/metabolism , Mice , Receptors, Notch/metabolism , Receptors, Notch/genetics , Endothelial Cells/metabolism , Endothelial Cells/cytology , Bile Ducts/cytology , Bile Ducts/metabolism , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/cytology , Transforming Growth Factor beta/metabolism
17.
Stem Cell Res Ther ; 15(1): 258, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39135132

ABSTRACT

BACKGROUND: Alzheimer disease (AD) is a heterogenous and multifactorial disease, and its pathology is partly driven by microglia and their activated phenotype. Brain organoids (BOs) are gaining prominence as a relevant model of the human brain for the study of AD; however, BOs are commonly devoid of microglia. To overcome this limitation, current protocols incorporate microglia through either (1) co-culture (BO co-culture), or (2) molecular manipulation at critical windows of BO development to have microglia arise innately (BO innate cultures). It is currently unclear whether the microglia incorporated into BOs by either of these two protocols differ in function. METHODS: At in vitro day 90, BO innate cultures and BO-co-cultures were challenged with the AD-related ß-amyloid peptide (Aß) for up to 72 h. After Aß challenge, BOs were collected for immunoblotting. Immunoblots compared immunodensity and protein banding of Aß and ionized calcium-binding adapter molecule 1 (IBA1, a marker of microglial activation) in BOs. The translational potential of these observations was supported using 56 human cortical samples from neurocognitively normal donors and patients with early-onset AD and late-onset AD. Statistical analyses were conducted using the Kruskal-Wallis test, a two-way ANOVA, or a simple linear regression, and where applicable, followed by Dunn's or Sidak's test. RESULTS: We show that BO co-cultures promote Aß oligomerization as early as 24 h and this coincides with a significant increase in IBA1 levels. In contrast, the Aßs do not oligomerize in BO innate cultures and the IBA1 response was modest and only emerged after 48 h. In human cortical samples, we found IBA1 levels correlated with age at onset, age at death, and the putative diagnostic Aß(1-42)/Aß(1-40) ratio (particularly in their oligomeric forms) in a sex-dependent manner. CONCLUSIONS: Our unique observations suggest that BOs with innate microglia model the response of a healthy brain to Aß, and by extension the initial stages of Aß challenge. It would be impossible to model these early stages of pathogenesis in BOs where microglia are already compromised, such as those with microglia incorporated by co-culture.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Brain , Coculture Techniques , Microglia , Organoids , Humans , Microglia/metabolism , Coculture Techniques/methods , Amyloid beta-Peptides/metabolism , Organoids/metabolism , Brain/metabolism , Brain/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Female , Male , Aged , Middle Aged
18.
J Vis Exp ; (209)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39141532

ABSTRACT

Due to the limited accessibility of the human retina, retinal organoids (ROs) are the best model for studying human retinal disease, which could reveal the mechanism of retinal development and the occurrence of retinal disease. Microglia (MG) are unique resident macrophages in the retina and central nervous system (CNS), serving crucial immunity functions. However, retinal organoids lack microglia since their differentiation origin is the yolk sac. The specific pathogenesis of microglia in these retinal diseases remains unclear; therefore, the establishment of a microglia-incorporated retinal organoid model turns out to be necessary. Here, we successfully constructed a co-cultured model of retinal organoids with microglia derived from human stem cells. In this article, we differentiated microglia and then co-cultured to retinal organoids in the early stage. As the incorporation of immune cells, this model provides an optimized platform for retinal disease modeling and drug screening to facilitate in-depth research on the pathogenesis and treatment of retinal and CNS-related diseases.


Subject(s)
Coculture Techniques , Microglia , Organoids , Retina , Organoids/cytology , Microglia/cytology , Retina/cytology , Humans , Coculture Techniques/methods , Cell Differentiation/physiology
19.
J Vis Exp ; (209)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39141550

ABSTRACT

Microglia are tissue-resident macrophages of the central nervous system (CNS), performing numerous functions that support neuronal health and CNS homeostasis. They are a major population of immune cells associated with CNS disease activity, adopting reactive phenotypes that potentially contribute to neuronal injury during chronic neurodegenerative diseases such as multiple sclerosis (MS). The distinct mechanisms by which microglia regulate neuronal function and survival during health and disease remain limited due to challenges in resolving the complex in vivo interactions between microglia, neurons, and other CNS environmental factors. Thus, the in vitro approach of co-culturing microglia and neurons remains a valuable tool for studying microglia-neuronal interactions. Here, we present a protocol to generate and co-culture primary microglia and neurons from mice. Specifically, microglia were isolated after 9-10 days in vitro from a mixed glia culture established from brain homogenates derived from neonatal mice between post-natal days 0-2. Neuronal cells were isolated from brain cortices of mouse embryos between embryonic days 16-18. After 4-5 days in vitro, neuronal cells were seeded in 96-well plates, followed by the addition of microglia to form the co-culture. Careful timing is critical for this protocol as both cell types need to reach experimental maturity to establish the co-culture. Overall, this co-culture can be useful for studying microglia-neuron interactions and can provide multiple readouts, including immunofluorescence microscopy, live imaging, as well as RNA and protein assays.


Subject(s)
Cerebral Cortex , Coculture Techniques , Microglia , Neurons , Animals , Coculture Techniques/methods , Microglia/cytology , Mice , Neurons/cytology , Cerebral Cortex/cytology , Cytological Techniques/methods
20.
Nat Commun ; 15(1): 7066, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39152123

ABSTRACT

DPANN is a widespread and diverse group of archaea characterized by their small size, reduced genome, limited metabolic pathways, and symbiotic existence. Known DPANN species are predominantly obligate ectosymbionts that depend on their host for proliferation. The structural and molecular details of host recognition, host-DPANN intercellular communication, and host adaptation in response to DPANN attachment remain unknown. Here, we use electron cryotomography (cryo-ET) to show that the Microcaldus variisymbioticus ARM-1 may interact with its host, Metallosphaera javensis AS-7 through intercellular proteinaceous nanotubes. Combining cryo-ET and sub-tomogram averaging, we show the in situ architectures of host and DPANN S-layers and the structures of the nanotubes in their primed and extended states. In addition, comparative proteomics and genomic analyses identified host proteomic changes in response to DPANN attachment. These results provide insights into the structural basis of host-DPANN communication and deepen our understanding of the host ectosymbiotic relationships.


Subject(s)
Cryoelectron Microscopy , Electron Microscope Tomography , Symbiosis , Electron Microscope Tomography/methods , Cryoelectron Microscopy/methods , Coculture Techniques/methods , Proteomics/methods , Archaeal Proteins/metabolism , Archaeal Proteins/genetics , Cell Communication , Archaea/metabolism , Archaea/genetics , Nanotubes/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL