Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.522
Filter
1.
Dev Cell ; 58(7): 616-632.e6, 2023 04 10.
Article in English | MEDLINE | ID: mdl-36990090

ABSTRACT

3D cell cultures, in particular organoids, are emerging models in the investigation of healthy or diseased tissues. Understanding the complex cellular sociology in organoids requires integration of imaging modalities across spatial and temporal scales. We present a multi-scale imaging approach that traverses millimeter-scale live-cell light microscopy to nanometer-scale volume electron microscopy by performing 3D cell cultures in a single carrier that is amenable to all imaging steps. This allows for following organoids' growth, probing their morphology with fluorescent markers, identifying areas of interest, and analyzing their 3D ultrastructure. We demonstrate this workflow on mouse and human 3D cultures and use automated image segmentation to annotate and quantitatively analyze subcellular structures in patient-derived colorectal cancer organoids. Our analyses identify local organization of diffraction-limited cell junctions in compact and polarized epithelia. The continuum-resolution imaging pipeline is thus suited to fostering basic and translational organoid research by simultaneously exploiting the advantages of light and electron microscopy.


Subject(s)
Cell Culture Techniques, Three Dimensional , Microscopy , Organoids , Animals , Humans , Mice , Cell Culture Techniques, Three Dimensional/methods , Microscopy, Electron , Organoids/diagnostic imaging , Organoids/physiology , Organoids/ultrastructure , Colorectal Neoplasms/pathology
2.
Cell Rep ; 38(7): 110379, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35172130

ABSTRACT

Pluripotent-stem-cell-derived human intestinal organoids (HIOs) model some aspects of intestinal development and disease, but current culture methods do not fully recapitulate the diverse cell types and complex organization of the human intestine and are reliant on 3D extracellular matrix or hydrogel systems, which limit experimental control and translational potential for regenerative medicine. We describe suspension culture as a simple, low-maintenance method for culturing HIOs and for promoting in vitro differentiation of an organized serosal mesothelial layer that is similar to primary human intestinal serosal mesothelium based on single-cell RNA sequencing and histological analysis. Functionally, HIO serosal mesothelium has the capacity to differentiate into smooth-muscle-like cells and exhibits fibrinolytic activity. An inhibitor screen identifies Hedgehog and WNT signaling as regulators of human serosal mesothelial differentiation. Collectively, suspension HIOs represent a three-dimensional model to study the human serosal mesothelium.


Subject(s)
Epithelium/growth & development , Intestines/growth & development , Organoids/growth & development , Serous Membrane/growth & development , Tissue Culture Techniques , Alginates/pharmacology , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cell Line , Collagen/pharmacology , Drug Combinations , Epithelium/drug effects , Hedgehog Proteins/metabolism , Humans , Intestines/ultrastructure , Laminin/pharmacology , Muscle, Smooth/cytology , Organoids/drug effects , Organoids/ultrastructure , Proteoglycans/pharmacology , Serous Membrane/drug effects , Serous Membrane/ultrastructure , Signal Transduction/drug effects , Suspensions , Wnt Proteins/metabolism
3.
Stem Cell Reports ; 16(11): 2690-2702, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34653402

ABSTRACT

Cases of Leber congenital amaurosis caused by mutations in CRX (LCA7) exhibit an early form of the disease and show signs of significant photoreceptor dysfunction and eventual loss. To establish a translational in vitro model system to study gene-editing-based therapies, we generated LCA7 retinal organoids harboring a dominant disease-causing mutation in CRX. Our LCA7 retinal organoids develop signs of immature and dysfunctional photoreceptor cells, providing us with a reliable in vitro model to recapitulate LCA7. Furthermore, we performed a proof-of-concept study in which we utilize allele-specific CRISPR/Cas9-based gene editing to knock out mutant CRX and saw moderate rescue of photoreceptor phenotypes in our organoids. This work provides early evidence for an effective approach to treat LCA7, which can be applied more broadly to other dominant genetic diseases.


Subject(s)
Gene Editing/methods , Genetic Predisposition to Disease/genetics , Homeodomain Proteins/genetics , Leber Congenital Amaurosis/genetics , Mutation , Trans-Activators/genetics , Alleles , Base Sequence , Cell Line , Gene Expression Profiling/methods , Genes, Dominant , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Leber Congenital Amaurosis/metabolism , Leber Congenital Amaurosis/pathology , Microscopy, Electron, Transmission , Models, Biological , Organoids/cytology , Organoids/metabolism , Organoids/ultrastructure , Phenotype , Polymorphism, Single Nucleotide , RNA-Seq/methods , Retina/metabolism , Trans-Activators/metabolism
4.
Elife ; 102021 10 26.
Article in English | MEDLINE | ID: mdl-34698018

ABSTRACT

During brain development, axons must extend over great distances in a relatively short amount of time. How the subcellular architecture of the growing axon sustains the requirements for such rapid build-up of cellular constituents has remained elusive. Human axons have been particularly poorly accessible to imaging at high resolution in a near-native context. Here, we present a method that combines cryo-correlative light microscopy and electron tomography with human cerebral organoid technology to visualize growing axon tracts. Our data reveal a wealth of structural details on the arrangement of macromolecules, cytoskeletal components, and organelles in elongating axon shafts. In particular, the intricate shape of the endoplasmic reticulum is consistent with its role in fulfilling the high demand for lipid biosynthesis to support growth. Furthermore, the scarcity of ribosomes within the growing shaft suggests limited translational competence during expansion of this compartment. These findings establish our approach as a powerful resource for investigating the ultrastructure of defined neuronal compartments.


Subject(s)
Axons/ultrastructure , Electron Microscope Tomography , Organoids/cytology , Brain/cytology , Brain/ultrastructure , Cryoelectron Microscopy , HeLa Cells , Humans , Macromolecular Substances/metabolism , Microscopy , Microscopy, Fluorescence , Organoids/ultrastructure
5.
Cells ; 10(7)2021 07 05.
Article in English | MEDLINE | ID: mdl-34359871

ABSTRACT

The creation of a testis organoid (artificial testis tissue) with sufficient resemblance to the complex form and function of the innate testis remains challenging, especially using non-rodent donor cells. Here, we report the generation of an organoid culture system with striking biomimicry of the native immature testis tissue, including vasculature. Using piglet testis cells as starting material, we optimized conditions for the formation of cell spheroids, followed by long-term culture in an air-liquid interface system. Both fresh and frozen-thawed cells were fully capable of self-reassembly into stable testis organoids consisting of tubular and interstitial compartments, with all major cell types and structural details expected in normal testis tissue. Surprisingly, our organoids also developed vascular structures; a phenomenon that has not been reported in any other culture system. In addition, germ cells do not decline over time, and Leydig cells release testosterone, hence providing a robust, tunable system for diverse basic and applied applications.


Subject(s)
Biomimetic Materials/pharmacology , Organoids/physiology , Testis/blood supply , Animals , Cell Count , Cryopreservation , Leydig Cells/cytology , Leydig Cells/drug effects , Luteinizing Hormone/metabolism , Male , Neovascularization, Physiologic/drug effects , Organ Specificity , Organoids/cytology , Organoids/drug effects , Organoids/ultrastructure , Swine , Testis/cytology , Testis/ultrastructure , Testosterone/metabolism
6.
BMC Mol Cell Biol ; 22(1): 37, 2021 Jul 05.
Article in English | MEDLINE | ID: mdl-34225662

ABSTRACT

BACKGROUND: Organ culture models have been used over the past few decades to study development and disease. The in vitro three-dimensional (3D) culture system of organoids is well known, however, these 3D systems are both costly and difficult to culture and maintain. As such, less expensive, faster and less complex methods to maintain 3D cell culture models would complement the use of organoids. Chick embryos have been used as a model to study human biology for centuries, with many fundamental discoveries as a result. These include cell type induction, cell competence, plasticity and contact inhibition, which indicates the relevance of using chick embryos when studying developmental biology and disease mechanisms. RESULTS: Here, we present an updated protocol that enables time efficient, cost effective and long-term expansion of fetal organ spheroids (FOSs) from chick embryos. Utilizing this protocol, we generated FOSs in an anchorage-independent growth pattern from seven different organs, including brain, lung, heart, liver, stomach, intestine and epidermis. These three-dimensional (3D) structures recapitulate many cellular and structural aspects of their in vivo counterpart organs and serve as a useful developmental model. In addition, we show a functional application of FOSs to analyze cell-cell interaction and cell invasion patterns as observed in cancer. CONCLUSION: The establishment of a broad ranging and highly effective method to generate FOSs from different organs was successful in terms of the formation of healthy, proliferating 3D organ spheroids that exhibited organ-like characteristics. Potential applications of chick FOSs are their use in studies of cell-to-cell contact, cell fusion and tumor invasion under defined conditions. Future studies will reveal whether chick FOSs also can be applicable in scientific areas such as viral infections, drug screening, cancer diagnostics and/or tissue engineering.


Subject(s)
Cell Culture Techniques, Three Dimensional , Models, Biological , Neoplasm Invasiveness/pathology , Organoids/cytology , Spheroids, Cellular/cytology , Animals , Cell Communication , Cell Line, Tumor , Chick Embryo , Chickens , Humans , Organoids/ultrastructure , Spheroids, Cellular/ultrastructure , Tissue Culture Techniques
7.
Cell Rep ; 36(3): 109351, 2021 07 20.
Article in English | MEDLINE | ID: mdl-34289360

ABSTRACT

Recurrence of uropathogenic Escherichia coli (UPEC) infections has been attributed to reactivation of quiescent intracellular reservoirs (QIRs) in deep layers of the bladder wall. QIRs are thought to arise late during infection following dispersal of bacteria from intracellular bacterial communities (IBCs) in superficial umbrella cells. Here, we track the formation of QIR-like bacteria in a bladder organoid model that recapitulates the stratified uroepithelium within a volume suitable for high-resolution live-cell imaging. Bacteria injected into the organoid lumen enter umbrella-like cells and proliferate to form IBC-like bodies. In parallel, single bacteria penetrate deeper layers of the organoid wall, where they localize within or between uroepithelial cells. These "solitary" bacteria evade killing by antibiotics and neutrophils and are morphologically distinct from bacteria in IBCs. We conclude that bacteria with QIR-like properties may arise at early stages of infection, independent of IBC formation and rupture.


Subject(s)
Anti-Bacterial Agents/pharmacology , Models, Biological , Neutrophils/pathology , Organoids/microbiology , Urinary Bladder/microbiology , Uropathogenic Escherichia coli/physiology , Animals , Cell Differentiation/drug effects , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Female , Humans , Imaging, Three-Dimensional , Mice, Inbred C57BL , Microbial Viability/drug effects , Movement , Neutrophils/drug effects , Organoids/drug effects , Organoids/ultrastructure , Urinary Bladder/pathology , Uropathogenic Escherichia coli/drug effects , Uropathogenic Escherichia coli/growth & development , Uropathogenic Escherichia coli/ultrastructure
8.
Cell ; 184(17): 4547-4563.e17, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34314701

ABSTRACT

Frontotemporal dementia (FTD) because of MAPT mutation causes pathological accumulation of tau and glutamatergic cortical neuronal death by unknown mechanisms. We used human induced pluripotent stem cell (iPSC)-derived cerebral organoids expressing tau-V337M and isogenic corrected controls to discover early alterations because of the mutation that precede neurodegeneration. At 2 months, mutant organoids show upregulated expression of MAPT, glutamatergic signaling pathways, and regulators, including the RNA-binding protein ELAVL4, and increased stress granules. Over the following 4 months, mutant organoids accumulate splicing changes, disruption of autophagy function, and build-up of tau and P-tau-S396. By 6 months, tau-V337M organoids show specific loss of glutamatergic neurons as seen in individuals with FTD. Mutant neurons are susceptible to glutamate toxicity, which can be rescued pharmacologically by the PIKFYVE kinase inhibitor apilimod. Our results demonstrate a sequence of events that precede neurodegeneration, revealing molecular pathways associated with glutamate signaling as potential targets for therapeutic intervention in FTD.


Subject(s)
Cerebrum/pathology , ELAV-Like Protein 4/genetics , Glutamic Acid/metabolism , Mutation/genetics , Neurons/pathology , Organoids/metabolism , RNA Splicing/genetics , tau Proteins/genetics , Autophagy/drug effects , Autophagy/genetics , Biomarkers/metabolism , Body Patterning/drug effects , Body Patterning/genetics , Cell Death/drug effects , Cell Line , Humans , Hydrazones/pharmacology , Lysosomes/drug effects , Lysosomes/metabolism , Morpholines/pharmacology , Neurons/drug effects , Neurons/metabolism , Organoids/drug effects , Organoids/ultrastructure , Phosphorylation/drug effects , Pyrimidines/pharmacology , RNA Splicing/drug effects , Signal Transduction/drug effects , Stress Granules/drug effects , Stress Granules/metabolism , Synapses/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics
9.
J Cell Biol ; 220(9)2021 09 06.
Article in English | MEDLINE | ID: mdl-34160561

ABSTRACT

Cells are 3D objects. Therefore, volume EM (vEM) is often crucial for correct interpretation of ultrastructural data. Today, scanning EM (SEM) methods such as focused ion beam (FIB)-SEM are frequently used for vEM analyses. While they allow automated data acquisition, precise targeting of volumes of interest within a large sample remains challenging. Here, we provide a workflow to target FIB-SEM acquisition of fluorescently labeled cells or subcellular structures with micrometer precision. The strategy relies on fluorescence preservation during sample preparation and targeted trimming guided by confocal maps of the fluorescence signal in the resin block. Laser branding is used to create landmarks on the block surface to position the FIB-SEM acquisition. Using this method, we acquired volumes of specific single cells within large tissues such as 3D cultures of mouse mammary gland organoids, tracheal terminal cells in Drosophila melanogaster larvae, and ovarian follicular cells in adult Drosophila, discovering ultrastructural details that could not be appreciated before.


Subject(s)
Drosophila melanogaster/ultrastructure , Granulosa Cells/ultrastructure , Mammary Glands, Animal/ultrastructure , Microscopy, Electron, Scanning/methods , Staining and Labeling/methods , Theca Cells/ultrastructure , Trachea/ultrastructure , Animals , Drosophila melanogaster/metabolism , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Female , Gene Expression , Genes, Reporter , Granulosa Cells/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Larva/metabolism , Larva/ultrastructure , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mammary Glands, Animal/metabolism , Mice , Microscopy, Electron, Scanning/instrumentation , Organoids/metabolism , Organoids/ultrastructure , Single-Cell Analysis/instrumentation , Single-Cell Analysis/methods , Theca Cells/metabolism , Trachea/metabolism , Workflow , Red Fluorescent Protein
10.
Dev Cell ; 56(9): 1346-1358.e6, 2021 05 03.
Article in English | MEDLINE | ID: mdl-33945785

ABSTRACT

Myelination is essential for central nervous system (CNS) formation, health, and function. Emerging evidence of oligodendrocyte heterogeneity in health and disease and divergent CNS gene expression profiles between mice and humans supports the development of experimentally tractable human myelination systems. Here, we developed human iPSC-derived myelinating organoids ("myelinoids") and quantitative tools to study myelination from oligodendrogenesis through to compact myelin formation and myelinated axon organization. Using patient-derived cells, we modeled a monogenetic disease of myelinated axons (Nfasc155 deficiency), recapitulating impaired paranodal axo-glial junction formation. We also validated the use of myelinoids for pharmacological assessment of myelination-both at the level of individual oligodendrocytes and globally across whole myelinoids-and demonstrated reduced myelination in response to suppressed synaptic vesicle release. Our study provides a platform to investigate human myelin development, disease, and adaptive myelination.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Myelin Sheath/physiology , Organoids/physiology , Axons/metabolism , Axons/ultrastructure , Humans , Myelin Sheath/ultrastructure , Nerve Growth Factors/deficiency , Nerve Growth Factors/metabolism , Organoids/ultrastructure , Tetanus Toxin/pharmacology , Time Factors
11.
Stem Cells Dev ; 30(8): 399-417, 2021 04.
Article in English | MEDLINE | ID: mdl-33677999

ABSTRACT

Progressive vision loss, caused by retinal degenerative (RD) diseases such as age-related macular degeneration, retinitis pigmentosa, and Leber congenital amaurosis, severely impacts quality of life and affects millions of people. Finding efficient treatment for blinding diseases is among the greatest unmet clinical needs. The evagination of optic vesicles from developing pluripotent stem cell-derived neuroepithelium and self-organization, lamination, and differentiation of retinal tissue in a dish generated considerable optimism for developing innovative approaches for treating RD diseases, which previously were not feasible. Retinal organoids may be a limitless source of multipotential retinal progenitors, photoreceptors (PRs), and the whole retinal tissue, which are productive approaches for developing RD disease therapies. In this study we compared the distribution and expression level of molecular markers (genetic and epigenetic) in human fetal retina (age 8-16 weeks) and human embryonic stem cell (hESC)-derived retinal tissue (organoids) by immunohistochemistry, RNA-seq, flow cytometry, and mass-spectrometry (to measure methylated and hydroxymethylated cytosine level), with a focus on PRs to evaluate the clinical application of hESC-retinal tissue for vision restoration. Our results revealed high correlation in gene expression profiles and histological profiles between human fetal retina (age 8-13 weeks) and hESC-derived retinal tissue (10-12 weeks). The transcriptome signature of hESC-derived retinal tissue from retinal organoids maintained for 24 weeks in culture resembled the transcriptome of human fetal retina of more advanced developmental stages. The histological profiles of 24 week-old hESC-derived retinal tissue displayed mature PR immunophenotypes and presence of developing inner and outer segments. Collectively, our work highlights the similarity of hESC-derived retinal tissue at early stages of development (10 weeks), and human fetal retina (age 8-13 weeks) and it supports the development of regenerative medicine therapies aimed at using tissue from hESC-derived retinal organoids (hESC-retinal implants) for mitigating vision loss.


Subject(s)
Cell Differentiation/genetics , Human Embryonic Stem Cells/metabolism , Organoids/metabolism , Pluripotent Stem Cells/metabolism , Retina/metabolism , Transcriptome/genetics , Cell Line , DNA Methylation , Homeodomain Proteins/metabolism , Human Embryonic Stem Cells/cytology , Humans , Immunohistochemistry , Ki-67 Antigen/metabolism , Microscopy, Confocal , Microscopy, Electron, Transmission , Organoids/cytology , Organoids/ultrastructure , PAX6 Transcription Factor/metabolism , Pluripotent Stem Cells/cytology , RNA-Seq/methods , Retina/cytology , Retina/embryology , Time Factors , Transcription Factors/metabolism
12.
Cells ; 10(2)2021 01 28.
Article in English | MEDLINE | ID: mdl-33525555

ABSTRACT

Humans with biallelic inactivating mutations in Epithelial Cell Adhesion Molecule (EpCAM) develop congenital tufting enteropathy (CTE). To gain mechanistic insights regarding EpCAM function in this disorder, we prepared intestinal epithelial cell (IEC) organoids and spheroids. IEC organoids and spheroids were generated from ROSA-CreERT2 EpCAMfl/fl mice. Proliferation, tight junctions, cell polarity and epithelial integrity were assessed in tamoxifen-induced EpCAM-deficient organoids via confocal immunofluorescence microscopy and Western blotting. Olfm4-expressing stem cells were assessed in IEC cells in vitro and in vivo via fluorescence in situ hybridization. To determine if existing drugs could ameliorate effects of EpCAM deficiency in IEC cells, a variety of pharmacologic inhibitors were screened. Deletion of EpCAM resulted in increased apoptosis and attenuated growth of organoids and spheroids. Selected claudins were destabilized and epithelial integrity was severely compromised. Epithelial integrity was improved by treatment with Rho-associated coiled-coil kinase (ROCK) inhibitors without restoration of claudin expression. Correspondingly, enhanced phosphorylation of myosin light chain, a serine/threonine ROCK substrate, was observed in EpCAM-deficient organoids. Strikingly, frequencies of Olfm4-expressing stem cells in EpCAM-deficient IEC cells in vitro and in vivo were decreased. Treatment with ROCK inhibitors increased numbers of stem cells in EpCAM-deficient organoids and spheroids. Thus, EpCAM regulates intestinal epithelial homeostasis via a signaling pathway that includes ROCK.


Subject(s)
Epithelial Cell Adhesion Molecule/metabolism , Epithelial Cells/metabolism , Intestines/cytology , Stem Cells/metabolism , rho-Associated Kinases/metabolism , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Polarity/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Claudins/metabolism , Epithelial Cells/drug effects , Gene Silencing , Intestinal Mucosa/metabolism , Mice, Knockout , Myosin Light Chains/metabolism , Organoids/drug effects , Organoids/metabolism , Organoids/ultrastructure , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Stem Cells/drug effects , rho-Associated Kinases/antagonists & inhibitors
13.
Sci Rep ; 11(1): 1944, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33479301

ABSTRACT

The poor prognosis of pancreatic ductal adenocarcinoma (PDAC) is attributed to the highly fibrotic stroma and complex multi-cellular microenvironment that is difficult to fully recapitulate in pre-clinical models. To fast-track translation of therapies and to inform personalised medicine, we aimed to develop a whole-tissue ex vivo explant model that maintains viability, 3D multicellular architecture, and microenvironmental cues of human pancreatic tumours. Patient-derived surgically-resected PDAC tissue was cut into 1-2 mm explants and cultured on gelatin sponges for 12 days. Immunohistochemistry revealed that human PDAC explants were viable for 12 days and maintained their original tumour, stromal and extracellular matrix architecture. As proof-of-principle, human PDAC explants were treated with Abraxane and we observed different levels of response between patients. PDAC explants were also transfected with polymeric nanoparticles + Cy5-siRNA and we observed abundant cytoplasmic distribution of Cy5-siRNA throughout the PDAC explants. Overall, our novel model retains the 3D architecture of human PDAC and has advantages over standard organoids: presence of functional multi-cellular stroma and fibrosis, and no tissue manipulation, digestion, or artificial propagation of organoids. This provides unprecedented opportunity to study PDAC biology including tumour-stromal interactions and rapidly assess therapeutic response to drive personalised treatment.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Pancreatic Ductal/genetics , Cell Culture Techniques , Organoids/pathology , Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Extracellular Matrix/pathology , Extracellular Matrix/ultrastructure , Humans , Organoids/ultrastructure , Pancreas/pathology , Pancreas/ultrastructure , Tumor Microenvironment/genetics
14.
Nat Protoc ; 16(1): 239-262, 2021 01.
Article in English | MEDLINE | ID: mdl-33247285

ABSTRACT

Advances in light-sheet and confocal microscopy now allow imaging of cleared large biological tissue samples and enable the 3D appreciation of cell and protein localization in their native organ environment. However, the sample preparations for such imaging are often onerous, and their capability for antigen detection is limited. Here, we describe FLASH (fast light-microscopic analysis of antibody-stained whole organs), a simple, rapid, fully customizable technique for molecular phenotyping of intact tissue volumes. FLASH utilizes non-degradative epitope recovery and membrane solubilization to enable the detection of a multitude of membranous, cytoplasmic and nuclear antigens in whole mouse organs and embryos, human biopsies, organoids and Drosophila. Retrieval and immunolabeling of epithelial markers, an obstacle for previous clearing techniques, can be achieved with FLASH. Upon volumetric imaging, FLASH-processed samples preserve their architecture and integrity and can be paraffin-embedded for subsequent histopathological analysis. The technique can be performed by scientists trained in light microscopy and yields results in <1 week.


Subject(s)
Antigens/analysis , Fluorescent Antibody Technique/methods , Imaging, Three-Dimensional/methods , Microscopy, Confocal/methods , Animals , Drosophila , Epitopes/analysis , Female , Humans , Kidney/ultrastructure , Lacrimal Apparatus/ultrastructure , Liver/ultrastructure , Lung/ultrastructure , Male , Mammary Glands, Human/ultrastructure , Mice , Organoids/ultrastructure , Pancreas/ultrastructure , Stomach/ultrastructure
15.
J Mol Med (Berl) ; 99(4): 449-462, 2021 04.
Article in English | MEDLINE | ID: mdl-33221939

ABSTRACT

Organoids constitute biological systems which are used to model organ development, homeostasis, regeneration, and disease in vitro and hold promise for use in therapy. Reflecting in vivo development, organoids form from tissue cells or pluripotent stem cells. Cues provided from the media and individual cells promote self-organization of these uniform starting cells into a structure, with emergent differentiated cells, morphology, and often functionality that resemble the tissue of origin. Therefore, organoids provide a complement to two-dimensional in vitro culture and in vivo animal models of development, providing the experimental control and flexibility of in vitro methods with the three-dimensional context of in vivo models, with fewer ethical restraints than human or animal work. However, using organoids, we are only just beginning to understand on the cellular level how the external conditions and signaling between individual cells promote the emergence of cells and structures. In this review, we focus specifically on organoids derived from endodermal tissues: the starting conditions of the cells, signaling mechanisms, and external media that allow the emergence of higher order self-organization.


Subject(s)
Endoderm/cytology , Organoids/cytology , Adult Stem Cells/cytology , Animals , Cell Communication , Cell Culture Techniques/methods , Cell Differentiation , Feedback, Physiological , Humans , Induced Pluripotent Stem Cells/cytology , Intestines/cytology , Mice , Morphogenesis , Organ Specificity , Organogenesis , Organoids/ultrastructure , Signal Transduction
16.
Cell ; 183(7): 1913-1929.e26, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33333020

ABSTRACT

Neurons in the cerebral cortex connect through descending pathways to hindbrain and spinal cord to activate muscle and generate movement. Although components of this pathway have been previously generated and studied in vitro, the assembly of this multi-synaptic circuit has not yet been achieved with human cells. Here, we derive organoids resembling the cerebral cortex or the hindbrain/spinal cord and assemble them with human skeletal muscle spheroids to generate 3D cortico-motor assembloids. Using rabies tracing, calcium imaging, and patch-clamp recordings, we show that corticofugal neurons project and connect with spinal spheroids, while spinal-derived motor neurons connect with muscle. Glutamate uncaging or optogenetic stimulation of cortical spheroids triggers robust contraction of 3D muscle, and assembloids are morphologically and functionally intact for up to 10 weeks post-fusion. Together, this system highlights the remarkable self-assembly capacity of 3D cultures to form functional circuits that could be used to understand development and disease.


Subject(s)
Cerebral Cortex/physiology , Motor Cortex/physiology , Organoids/physiology , Animals , Calcium/metabolism , Cell Differentiation , Cells, Cultured , Cervical Vertebrae , Gene Expression Regulation , Glutamates/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Mice , Muscles/physiology , Myoblasts/metabolism , Nerve Net/physiology , Optogenetics , Organoids/ultrastructure , Rhombencephalon/physiology , Spheroids, Cellular/cytology , Spinal Cord/cytology
17.
Sci Rep ; 10(1): 20937, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33262363

ABSTRACT

The endoplasmic reticulum (ER) is a complex subcellular organelle composed of diverse structures such as tubules, sheets and tubular matrices. Flaviviruses such as Zika virus (ZIKV) induce reorganization of ER membranes to facilitate viral replication. Here, using 3D super resolution microscopy, ZIKV infection is shown to induce the formation of dense tubular matrices associated with viral replication in the central ER. Viral non-structural proteins NS4B and NS2B associate with replication complexes within the ZIKV-induced tubular matrix and exhibit distinct ER distributions outside this central ER region. Deep neural networks trained to distinguish ZIKV-infected versus mock-infected cells successfully identified ZIKV-induced central ER tubular matrices as a determinant of viral infection. Super resolution microscopy and deep learning are therefore able to identify and localize morphological features of the ER and allow for better understanding of how ER morphology changes due to viral infection.


Subject(s)
Deep Learning , Endoplasmic Reticulum/metabolism , Microscopy/methods , Zika Virus/physiology , Brain/pathology , Brain/virology , Cell Line, Tumor , Endoplasmic Reticulum/ultrastructure , Extracellular Matrix/metabolism , Humans , Organoids/metabolism , Organoids/ultrastructure , Organoids/virology , RNA, Double-Stranded/metabolism , Viral Nonstructural Proteins/metabolism , Zika Virus/ultrastructure , Zika Virus Infection/virology
18.
Sci Rep ; 10(1): 20292, 2020 11 20.
Article in English | MEDLINE | ID: mdl-33219246

ABSTRACT

To elucidate molecular pharmacology of Rho-associated coiled-coil containing protein kinase inhibitors (ROCK-i, Ripasudil and Y27632) on their efficiency for aqueous outflow, 2D or 3D cultures of a human trabecular meshwork (HTM) were prepared in the presence of TGFß2. Those were examined by transendothelial electrical resistance (TEER, 2D), electronic microscopy (EM, 2D and 3D), expression of the extracellular matrix (ECM) including collagen1 (COL1), COL4 and COL6, and fibronectin (FN) by immunolabeling and/or quantitative PCR (3D), and solidity of 3D organoids by a micro-squeezer. TGFß2 significantly increased the TEER values in 2D cultures, and the ECM expression indicated that the 3D organoids assumed a more densely packed shape. ROCK-i greatly reduced the TGFß2-induced enhancement of TEER and the immunolabeled ECM expression of the 3D organoids. In contrast, the mRNA expression of COL1 was increased, and those of COL4 and FN were unchanged. EM revealed that TGFß2 caused the HTM cells to become more compact and abundant ECM deposits within the 3D organoids were observed. These were significantly inhibited by ROCK-i. The dense solids caused by the presence of TGFß2 were significantly suppressed by ROCK-i. Current study indicates that ROCK-i cause beneficial effects toward the spatial configuration of TGFß2-induced HTM 3D organoids.


Subject(s)
Antihypertensive Agents/pharmacology , Glaucoma, Open-Angle/drug therapy , Optic Nerve Diseases/prevention & control , Trabecular Meshwork/drug effects , rho-Associated Kinases/antagonists & inhibitors , Amides/pharmacology , Amides/therapeutic use , Antihypertensive Agents/therapeutic use , Cell Culture Techniques , Cell Line , Glaucoma, Open-Angle/complications , Humans , Intraocular Pressure/drug effects , Isoquinolines/pharmacology , Isoquinolines/therapeutic use , Microscopy, Electron, Scanning , Optic Nerve Diseases/etiology , Organoids/drug effects , Organoids/metabolism , Organoids/ultrastructure , Pyridines/pharmacology , Pyridines/therapeutic use , Spheroids, Cellular , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Trabecular Meshwork/cytology , Trabecular Meshwork/metabolism , Trabecular Meshwork/ultrastructure , Transforming Growth Factor beta2/metabolism , rho-Associated Kinases/metabolism
19.
Nat Med ; 26(12): 1888-1898, 2020 12.
Article in English | MEDLINE | ID: mdl-32989314

ABSTRACT

22q11.2 deletion syndrome (22q11DS) is a highly penetrant and common genetic cause of neuropsychiatric disease. Here we generated induced pluripotent stem cells from 15 individuals with 22q11DS and 15 control individuals and differentiated them into three-dimensional (3D) cerebral cortical organoids. Transcriptional profiling across 100 days showed high reliability of differentiation and revealed changes in neuronal excitability-related genes. Using electrophysiology and live imaging, we identified defects in spontaneous neuronal activity and calcium signaling in both organoid- and 2D-derived cortical neurons. The calcium deficit was related to resting membrane potential changes that led to abnormal inactivation of voltage-gated calcium channels. Heterozygous loss of DGCR8 recapitulated the excitability and calcium phenotypes and its overexpression rescued these defects. Moreover, the 22q11DS calcium abnormality could also be restored by application of antipsychotics. Taken together, our study illustrates how stem cell derived models can be used to uncover and rescue cellular phenotypes associated with genetic forms of neuropsychiatric disease.


Subject(s)
Calcium Signaling/genetics , Cerebral Cortex/ultrastructure , DiGeorge Syndrome/diagnosis , Neurons/ultrastructure , Adult , Cell Differentiation/genetics , Cerebral Cortex/pathology , DiGeorge Syndrome/pathology , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/ultrastructure , Male , Neurons/pathology , Organoids/pathology , Organoids/ultrastructure , Young Adult
20.
Cell ; 182(6): 1623-1640.e34, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32946783

ABSTRACT

Human organoids recapitulating the cell-type diversity and function of their target organ are valuable for basic and translational research. We developed light-sensitive human retinal organoids with multiple nuclear and synaptic layers and functional synapses. We sequenced the RNA of 285,441 single cells from these organoids at seven developmental time points and from the periphery, fovea, pigment epithelium and choroid of light-responsive adult human retinas, and performed histochemistry. Cell types in organoids matured in vitro to a stable "developed" state at a rate similar to human retina development in vivo. Transcriptomes of organoid cell types converged toward the transcriptomes of adult peripheral retinal cell types. Expression of disease-associated genes was cell-type-specific in adult retina, and cell-type specificity was retained in organoids. We implicate unexpected cell types in diseases such as macular degeneration. This resource identifies cellular targets for studying disease mechanisms in organoids and for targeted repair in human retinas.


Subject(s)
Cell Differentiation/genetics , Organoids/cytology , Organoids/metabolism , Retina/cytology , Retina/metabolism , Single-Cell Analysis/methods , Synapses/physiology , Transcriptome/genetics , Cell Culture Techniques/methods , Cell Line , Electrophysiology , Female , Gene Expression Regulation, Developmental/genetics , Genetic Predisposition to Disease/genetics , Humans , In Situ Hybridization , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Microscopy, Electron , Multigene Family , Naphthoquinones , Organoids/radiation effects , Organoids/ultrastructure , Retina/pathology , Retina/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...