Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 774
Filter
1.
Ceska Gynekol ; 89(2): 151-155, 2024.
Article in English | MEDLINE | ID: mdl-38704229

ABSTRACT

The human placenta serves as a vital barrier between the mother and the developing fetus during pregnancy. A defect in the early development of the placenta is associated with severe pregnancy disorders. Despite its complex development, various molecular processes control placental development, and the specialization of trophoblast cells is still not fully understood. One primary obstacle is the lack of suitable cell model systems. Traditional two-dimensional (2D) cell cultures fail to mimic in vivo conditions and do not capture the intricate intercellular interactions vital for studying placental development. However, three-dimensional (3D) organoid models derived from stem cells that replicate natural cell organization and architecture have greatly improved our understanding of trophoblast behavior and its medicinal applications. Organoids with relevant phenotypes provide a valuable platform to model both placental physiology and pathology, including the modeling of placental disorders. They hold great promise for personalized medicine, improved diagnostics, and the evaluation of pharmaceutical drug efficacy and safety. This article provides a concise overview of trophoblast stem cells, trophoblast invasion, and the evolving role of organoids in gynecology.


Subject(s)
Organoids , Stem Cells , Trophoblasts , Humans , Trophoblasts/physiology , Organoids/physiology , Female , Pregnancy , Stem Cells/physiology , Placenta/cytology , Placenta/physiology , Placenta/pathology , Placentation/physiology
2.
Nat Commun ; 15(1): 2945, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38600094

ABSTRACT

An inter-regional cortical tract is one of the most fundamental architectural motifs that integrates neural circuits to orchestrate and generate complex functions of the human brain. To understand the mechanistic significance of inter-regional projections on development of neural circuits, we investigated an in vitro neural tissue model for inter-regional connections, in which two cerebral organoids are connected with a bundle of reciprocally extended axons. The connected organoids produced more complex and intense oscillatory activity than conventional or directly fused cerebral organoids, suggesting the inter-organoid axonal connections enhance and support the complex network activity. In addition, optogenetic stimulation of the inter-organoid axon bundles could entrain the activity of the organoids and induce robust short-term plasticity of the macroscopic circuit. These results demonstrated that the projection axons could serve as a structural hub that boosts functionality of the organoid-circuits. This model could contribute to further investigation on development and functions of macroscopic neuronal circuits in vitro.


Subject(s)
Axons , Neurons , Humans , Axons/physiology , Neurons/physiology , Organoids/physiology , Brain
3.
PLoS One ; 19(3): e0301079, 2024.
Article in English | MEDLINE | ID: mdl-38512940

ABSTRACT

Developing precise species-specific in vitro models that closely resemble in vivo intestinal tissues is essential for advancing our understanding of gastrointestinal physiology and associated diseases. This is especially crucial in examining host-pathogen interactions, particularly in bovines, a known reservoir for microbes and pathogens posing substantial public health threats. This research investigated the viability of producing bovine rectal organoids from cryopreserved tissues. We compared two cryopreservation methods with a traditional technique using fresh tissues, evaluating their effectiveness through growth rates, long-term viability, and comprehensive structural, cellular, and genetic analyses. These assessments utilized phase-contrast imaging, immunofluorescence imaging, and RT-qPCR assays. Additionally, the study developed a sophisticated method for forming a functional epithelial barrier from organoid-derived bovine rectal monolayers, incorporating a wide range of epithelial cells. This methodology employed transepithelial electrical resistance (TEER), parallel artificial membrane permeability assay (Papp), confocal microscopy, and advanced imaging techniques like scanning electron microscopy (SEM) and transmission electron microscopy (TEM). Our findings decisively show that bovine rectal organoids can be effectively generated from cryopreserved biopsy tissues. Moreover, we formulated a robust and optimized protocol for creating functional rectal monolayers from these organoids. This significant progress is particularly relevant given the susceptibility of the bovine rectum to various enteric pathogens of public health concern, marking a vital step forward in veterinary and biomedical research. The creation of accurate species specific in vitro models that faithfully mimic in vivo intestinal tissues is critical for enhancing our understanding of gut physiology and related pathologies. This is particularly relevant in studying the interactions between hosts and microbes or pathogens with significant public health risks where bovine can be the major reservoir.


Subject(s)
Cryopreservation , Rectum , Animals , Cattle , Epithelial Cells , Biopsy , Organoids/physiology , Intestinal Mucosa
5.
Commun Biol ; 7(1): 218, 2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38388772

ABSTRACT

Painted turtles are remarkable for their freeze tolerance and supercooling ability along with their associated resilience to hypoxia/anoxia and oxidative stress, rendering them an ideal biomedical model for hypoxia-induced injuries (including strokes), tissue cooling during surgeries, and organ cryopreservation. Yet, such research is hindered by their seasonal reproduction and slow maturation. Here we developed and characterized adult stem cell-derived turtle liver organoids (3D self-assembled in vitro structures) from painted, snapping, and spiny softshell turtles spanning ~175My of evolution, with a subset cryopreserved. This development is, to the best of our knowledge, a first for this vertebrate Order, and complements the only other non-avian reptile organoids from snake venom glands. Preliminary characterization, including morphological, transcriptomic, and proteomic analyses, revealed organoids enriched in cholangiocytes. Deriving organoids from distant turtles and life stages demonstrates that our techniques are broadly applicable to chelonians, permitting the development of functional genomic tools currently lacking in herpetological research. Such platform could potentially support studies including genome-to-phenome mapping, gene function, genome architecture, and adaptive responses to climate change, with implications for ecological, evolutionary, and biomedical research.


Subject(s)
Liver , Organoids , Turtles , Animals , Genome , Hypoxia/genetics , Proteomics , Turtles/physiology , Organoids/physiology
6.
Cell Rep Methods ; 4(1): 100686, 2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38218190

ABSTRACT

Precise modulation of brain activity is fundamental for the proper establishment and maturation of the cerebral cortex. To this end, cortical organoids are promising tools to study circuit formation and the underpinnings of neurodevelopmental disease. However, the ability to manipulate neuronal activity with high temporal resolution in brain organoids remains limited. To overcome this challenge, we introduce a bioelectronic approach to control cortical organoid activity with the selective delivery of ions and neurotransmitters. Using this approach, we sequentially increased and decreased neuronal activity in brain organoids with the bioelectronic delivery of potassium ions (K+) and γ-aminobutyric acid (GABA), respectively, while simultaneously monitoring network activity. This works highlights bioelectronic ion pumps as tools for high-resolution temporal control of brain organoid activity toward precise pharmacological studies that can improve our understanding of neuronal function.


Subject(s)
Cerebral Cortex , Neurons , Neurons/physiology , Organoids/physiology , Brain , Neurotransmitter Agents
7.
Eur J Immunol ; 54(2): e2250248, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37957831

ABSTRACT

Human intestinal organoids are an ideal model system for studying gastrointestinal physiology and immunopathology. Altered physiology and mucosal immune response are hallmarks of numerous intestinal functional and inflammatory diseases, including inflammatory bowel disease (IBD), coeliac disease, irritable bowel syndrome (IBS), and obesity. These conditions impact the normal epithelial functions of the intestine, such as absorption, barrier function, secretion, and host-microbiome communication. They are accompanied by characteristic intestinal symptoms and have significant societal, economic, and healthcare burdens. To develop new treatment options, cutting-edge research is required to investigate their etiology and pathology. Human intestinal organoids derived from patient tissue recapitulate the key physiological and immunopathological aspects of these conditions, providing a promising platform for elucidating disease mechanisms. This review will summarize recent reports on patient-derived human small intestinal and colonic organoids and highlight how these models have been used to study intestinal epithelial functions in the context of inflammation, altered physiology, and immune response. Furthermore, it will elaborate on the various organoid systems in use and the techniques/assays currently available to study epithelial functions. Finally, it will conclude by discussing the limitations and future perspectives of organoid technology.


Subject(s)
Inflammatory Bowel Diseases , Intestinal Mucosa , Humans , Intestines/pathology , Organoids/pathology , Organoids/physiology , Intestine, Small/pathology
8.
Cell Mol Life Sci ; 80(11): 318, 2023 Oct 07.
Article in English | MEDLINE | ID: mdl-37804439

ABSTRACT

Our current knowledge regarding the development of the human brain mostly derives from experimental studies on non-human primates, sheep, and rodents. However, these studies may not completely simulate all the features of human brain development as a result of species differences and variations in pre- and postnatal brain maturation. Therefore, it is important to supplement the in vivo animal models to increase the possibility that preclinical studies have appropriate relevance for potential future human trials. Three-dimensional brain organoid culture technology could complement in vivo animal studies to enhance the translatability of the preclinical animal studies and the understanding of brain-related disorders. In this review, we focus on the development of a model of hypoxic-ischemic (HI) brain injury using human brain organoids to complement the translation from animal experiments to human pathophysiology. We also discuss how the development of these tools provides potential opportunities to study fundamental aspects of the pathophysiology of HI-related brain injury including differences in the responses between males and females.


Subject(s)
Brain Injuries , Hypoxia-Ischemia, Brain , Male , Female , Animals , Humans , Sheep , Disease Models, Animal , Brain , Rodentia , Organoids/physiology
9.
Zhonghua Jie He He Hu Xi Za Zhi ; 46(8): 829-834, 2023 Aug 12.
Article in Chinese | MEDLINE | ID: mdl-37536996

ABSTRACT

Organoids are tissue cultures formed by culturing cells in three-dimensional environments that simulate the physiological or pathological conditions of the human body. The cultivation of organoids is used to study the temporal and spatial transformation of cells during the development of tissues or organs, to investigate changes in cellular functions and inter-communications caused by various risk factors, and to discover potential therapeutic targets. This article provided an overview of the cultivation and identification methods of alveolar organoids, as well as the research progress in their application to common respiratory diseases such as pulmonary fibrosis, chronic obstructive pulmonary disease, viral pneumonia, and so on. The limitations and future applications of alveolar organoids are also analyzed and discussed.


Subject(s)
Lung Diseases , Pneumonia, Viral , Pulmonary Disease, Chronic Obstructive , Humans , Lung/pathology , Lung Diseases/pathology , Pneumonia, Viral/pathology , Organoids/pathology , Organoids/physiology , Pulmonary Disease, Chronic Obstructive/pathology
10.
PLoS Comput Biol ; 19(8): e1011386, 2023 08.
Article in English | MEDLINE | ID: mdl-37578984

ABSTRACT

Organoids offer a powerful model to study cellular self-organisation, the growth of specific tissue morphologies in-vitro, and to assess potential medical therapies. However, the intrinsic mechanisms of these systems are not entirely understood yet, which can result in variability of organoids due to differences in culture conditions and basement membrane extracts used. Improving the standardisation of organoid cultures is essential for their implementation in clinical protocols. Developing tools to assess and predict the behaviour of these systems may produce a more robust and standardised biological model to perform accurate clinical studies. Here, we developed an algorithm to automate crypt-like structure counting on intestinal organoids in both in-vitro and in-silico images. In addition, we modified an existing two-dimensional agent-based mathematical model of intestinal organoids to better describe the system physiology, and evaluated its ability to replicate budding structures compared to new experimental data we generated. The crypt-counting algorithm proved useful in approximating the average number of budding structures found in our in-vitro intestinal organoid culture images on days 3 and 7 after seeding. Our changes to the in-silico model maintain the potential to produce simulations that replicate the number of budding structures found on days 5 and 7 of in-vitro data. The present study aims to aid in quantifying key morphological structures and provide a method to compare both in-vitro and in-silico experiments. Our results could be extended later to 3D in-silico models.


Subject(s)
Intestines , Stem Cells , Computer Simulation , Organoids/physiology , Intestinal Mucosa
11.
Handb Exp Pharmacol ; 281: 59-81, 2023.
Article in English | MEDLINE | ID: mdl-37306818

ABSTRACT

Induced pluripotent stem cells (iPSCs) can be differentiated into specific neurons and brain organoids by adding induction factors and small molecules in vitro, which carry human genetic information and recapitulate the development process of human brain as well as physiological, pathological, and pharmacological characteristics. Hence, iPSC-derived neurons and organoids hold great promise for studying human brain development and related nervous system diseases in vitro, and provide a platform for drug screening. In this chapter, we summarize the development of the differentiation techniques for neurons and brain organoids from iPSCs, and their applications in studying brain disease, drug screening, and transplantation.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Induced Pluripotent Stem Cells/physiology , Brain , Neurons , Cell Differentiation , Organoids/physiology
12.
Endokrynol Pol ; 74(2): 121-127, 2023.
Article in English | MEDLINE | ID: mdl-37039493

ABSTRACT

Organoids are derived from stem cells under three-dimensional culture conditions through self-assembly, and they can recapitulate the structural and functional characteristics of organs in vivo during culture. Organoids can be generated from both normal and malignant tissues. Those derived from normal tissues are widely used in the field of regenerative medicine. Meanwhile, tumour-derived organoids retain the phenotypic heterogeneity and atypia of the primary tumour, thereby providing a reliable in vitro model for the study of tumour pathogenesis and treatment. The thyroid gland is one of the most important endocrine organs regulating the body's energy metabolism and growth; however, it is also associated with a high incidence of malignancy. Organoid is an effective tool for thyroid research. Thyroid tumour-derived organoids can inherit the histopathological properties of primary tumours, and thyroid tissue-derived organoids can form follicular structures and secrete thyroid hormones. The above characteristics of organoids provide a reliable way to study the mechanism of thyroid genesis and tumour development in vitro. In this review, we focus on current knowledge and strategies for the establishment of thyroid organoids in thyroid regeneration and tumour research aiming to increase our understanding of the pathogenesis of thyroid tumours and the regenerative treatment of patients with hypothyroidism.


Subject(s)
Organoids , Thyroid Neoplasms , Humans , Organoids/pathology , Organoids/physiology , Regenerative Medicine , Thyroid Neoplasms/pathology
13.
Nature ; 616(7955): 143-151, 2023 04.
Article in English | MEDLINE | ID: mdl-36991123

ABSTRACT

The relationship between the human placenta-the extraembryonic organ made by the fetus, and the decidua-the mucosal layer of the uterus, is essential to nurture and protect the fetus during pregnancy. Extravillous trophoblast cells (EVTs) derived from placental villi infiltrate the decidua, transforming the maternal arteries into high-conductance vessels1. Defects in trophoblast invasion and arterial transformation established during early pregnancy underlie common pregnancy disorders such as pre-eclampsia2. Here we have generated a spatially resolved multiomics single-cell atlas of the entire human maternal-fetal interface including the myometrium, which enables us to resolve the full trajectory of trophoblast differentiation. We have used this cellular map to infer the possible transcription factors mediating EVT invasion and show that they are preserved in in vitro models of EVT differentiation from primary trophoblast organoids3,4 and trophoblast stem cells5. We define the transcriptomes of the final cell states of trophoblast invasion: placental bed giant cells (fused multinucleated EVTs) and endovascular EVTs (which form plugs inside the maternal arteries). We predict the cell-cell communication events contributing to trophoblast invasion and placental bed giant cell formation, and model the dual role of interstitial EVTs and endovascular EVTs in mediating arterial transformation during early pregnancy. Together, our data provide a comprehensive analysis of postimplantation trophoblast differentiation that can be used to inform the design of experimental models of the human placenta in early pregnancy.


Subject(s)
Multiomics , Pregnancy Trimester, First , Trophoblasts , Female , Humans , Pregnancy , Cell Movement , Placenta/blood supply , Placenta/cytology , Placenta/physiology , Pregnancy Trimester, First/physiology , Trophoblasts/cytology , Trophoblasts/metabolism , Trophoblasts/physiology , Decidua/blood supply , Decidua/cytology , Maternal-Fetal Relations/physiology , Single-Cell Analysis , Myometrium/cytology , Myometrium/physiology , Cell Differentiation , Organoids/cytology , Organoids/physiology , Stem Cells/cytology , Transcriptome , Transcription Factors/metabolism , Cell Communication
14.
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
15.
Hist Philos Life Sci ; 45(2): 14, 2023 Mar 22.
Article in English | MEDLINE | ID: mdl-36949354

ABSTRACT

Organoids and organs-on-a-chip are currently the two major families of 3D advanced organotypic in vitro culture systems, aimed at reconstituting miniaturized models of physiological and pathological states of human organs. Both share the tenets of the so-called "three-dimensional thinking", a Systems Physiology approach focused on recapitulating the dynamic interactions between cells and their microenvironment. We first review the arguments underlying the "paradigm shift" toward three-dimensional thinking in the in vitro culture community. Then, through a historically informed account of the technical affordances and the epistemic commitments of these two approaches, we highlight how they embody two distinct experimental cultures. We finally argue that the current systematic effort for their integration requires not only innovative "synergistic" engineering solutions, but also conceptual integration between different perspectives on biological causality.


Subject(s)
Organoids , Tissue Engineering , Humans , Tissue Engineering/methods , Organoids/physiology , Microphysiological Systems , Philosophy
16.
Hepatol Commun ; 7(4)2023 04 01.
Article in English | MEDLINE | ID: mdl-36972388

ABSTRACT

In the past decade, liver organoids have evolved rapidly as valuable research tools, providing novel insights into almost all types of liver diseases, including monogenic liver diseases, alcohol-associated liver disease, metabolic-associated fatty liver disease, various types of (viral) hepatitis, and liver cancers. Liver organoids in part mimic the microphysiology of the human liver and fill a gap in high-fidelity liver disease models to a certain extent. They hold great promise to elucidate the pathogenic mechanism of a diversity of liver diseases and play a crucial role in drug development. Moreover, it is challenging but opportunistic to apply liver organoids for tailored therapies of various liver diseases. The establishment, applications, and challenges of different types of liver organoids, for example, derived from embryonic, adult, or induced pluripotent stem cells, to model different liver diseases, are presented in this review.


Subject(s)
Induced Pluripotent Stem Cells , Liver Neoplasms , Adult , Humans , Induced Pluripotent Stem Cells/pathology , Liver Neoplasms/pathology , Drug Development , Organoids/physiology
17.
Biol Psychiatry ; 93(7): 632-641, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36739210

ABSTRACT

Neuropsychiatric research has been impeded by limited access to human brain tissue, especially from early stages of neurodevelopment when the pathophysiology of many childhood-onset disorders is initiated. Neural organoids are 3-dimensional, self-organizing, multicellular structures generated from pluripotent stem cells that recapitulate some of the cell diversity, cytoarchitecture, and functional features of domains of the developing nervous system. Assembloids are 3-dimensional, self-organizing cultures created by the combination of two or more distinctly patterned organoids or an organoid plus additional cell or tissue type(s) that are used to model cell migration and connectivity. Here we review recent advances in neuropsychiatric disorder research using organoid and assembloid models to study the role of disease-relevant genes and mutations, as well as the impact of environmental risk factors on neural development. We also highlight some of the advantages and limitations of these model systems in bringing insights into the pathophysiology of neuropsychiatric disorders.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Child , Brain/physiology , Organoids/physiology , Models, Biological
18.
AJOB Neurosci ; 14(2): 178-196, 2023.
Article in English | MEDLINE | ID: mdl-36794285

ABSTRACT

Human cerebral organoids are three-dimensional biological cultures grown in the laboratory to mimic as closely as possible the cellular composition, structure, and function of the corresponding organ, the brain. For now, cerebral organoids lack blood vessels and other characteristics of the human brain, but are also capable of having coordinated electrical activity. They have been usefully employed for the study of several diseases and the development of the nervous system in unprecedented ways. Research on human cerebral organoids is proceeding at a very fast pace and their complexity is bound to improve. This raises the question of whether cerebral organoids will also be able to develop the unique feature of the human brain, consciousness. If this is the case, some ethical issues would arise. In this article, we discuss the necessary neural correlates and constraints for the emergence of consciousness according to some of the most debated neuroscientific theories. Based on this, we consider what the moral status of a potentially conscious brain organoid might be, in light of ethical and ontological arguments. We conclude by proposing a precautionary principle and some leads for further investigation. In particular, we consider the outcomes of some very recent experiments as entities of a potential new kind.


Subject(s)
Consciousness , Nervous System Physiological Phenomena , Humans , Consciousness/physiology , Brain , Moral Status , Organoids/physiology
19.
Cells Tissues Organs ; 212(5): 369-382, 2023.
Article in English | MEDLINE | ID: mdl-36716724

ABSTRACT

Valid and relevant models are critical for research to have biological relevance or to proceed in the right path. As well-established two-dimensional cell cultures lack niches and cues and rodent models differ in species, three-dimensional organoids emerged as a powerful platform for research. Cultured in vitro from stem cells, organoids are heterogeneous in cells and closely resemble the in vivo settings. Organoids also recapitulate the unique human features if cultured from a human source and are subjected to genetic modification. However, one type of organoid possesses only a limited selection of cells. In particular, the absence of vasculature and immune cells restricts the organoids from nutrition, cues, or critical interactions, undermining the validity of organoids as physiological or pathological models. To fill the current gap, there is an urgent need to provide organoids with vasculature and immune cells. In this paper, we review the methods to generate physiological and pathological organoid models and summarize ways to vascularize or immunize them. Our discussion continues with some advantages and disadvantages of each method and some emerging solutions to current problems.


Subject(s)
Cell Culture Techniques , Organoids , Humans , Organoids/physiology , Stem Cells
20.
Semin Reprod Med ; 41(5): 160-171, 2023 Sep.
Article in English | MEDLINE | ID: mdl-38262440

ABSTRACT

The female reproductive tract (FRT) is home to diverse microbial communities that play a pivotal role in reproductive health and disorders such as infertility, endometriosis, and cervical cancer. To understand the complex host-microbiota interactions within the FRT, models that authentically replicate the FRT's environment, including the interplay between the microbiota, mucus layer, immune system, and hormonal cycle, are key. Recent strides in organoid and microfluidic technologies are propelling research in this domain, offering insights into FRT-microbiota interactions and potential therapeutic avenues. This review delves into the current state of FRT organoid models and microbe integration techniques, evaluating their merits and challenges for specific research objectives. Emphasis is placed on innovative approaches and applications, including integrating organoids with microfluidics, and using patient-derived biobanks, as this offers potential for deeper mechanistic insights and personalized therapeutic strategies. Modeling various FRT properties in organoids is explored, from encompassing age-related epithelial features, oxygen levels, and hormonal effects to mucus layers, immune responses, and microbial interactions, highlighting their potential to transform reproductive health research and predict possible outcomes.


Subject(s)
Endometriosis , Microbiota , Female , Humans , Microphysiological Systems , Genitalia, Female , Microbiota/physiology , Organoids/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...