Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 321
Filtrar
1.
PLoS Comput Biol ; 20(8): e1012256, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39093897

RESUMO

Patient-derived tumor organoids (PDTOs) are novel cellular models that maintain the genetic, phenotypic and structural features of patient tumor tissue and are useful for studying tumorigenesis and drug response. When integrated with advanced 3D imaging and analysis techniques, PDTOs can be used to establish physiologically relevant high-throughput and high-content drug screening platforms that support the development of patient-specific treatment strategies. However, in order to effectively leverage high-throughput PDTO observations for clinical predictions, it is critical to establish a quantitative understanding of the basic properties and variability of organoid growth dynamics. In this work, we introduced an innovative workflow for analyzing and understanding PDTO growth dynamics, by integrating a high-throughput imaging deep learning platform with mathematical modeling, incorporating flexible growth laws and variable dormancy times. We applied the workflow to colon cancer organoids and demonstrated that organoid growth is well-described by the Gompertz model of growth. Our analysis showed significant intrapatient heterogeneity in PDTO growth dynamics, with the initial exponential growth rate of an organoid following a lognormal distribution within each dataset. The level of intrapatient heterogeneity varied between patients, as did organoid growth rates and dormancy times of single seeded cells. Our work contributes to an emerging understanding of the basic growth characteristics of PDTOs, and it highlights the heterogeneity in organoid growth both within and between patients. These results pave the way for further modeling efforts aimed at predicting treatment response dynamics and drug resistance timing.


Assuntos
Organoides , Humanos , Organoides/crescimento & desenvolvimento , Organoides/efeitos dos fármacos , Organoides/patologia , Modelos Biológicos , Neoplasias do Colo/diagnóstico por imagem , Neoplasias do Colo/patologia , Neoplasias do Colo/tratamento farmacológico , Biologia Computacional , Aprendizado Profundo , Modelos Teóricos , Imageamento Tridimensional/métodos
2.
Differentiation ; 137: 100781, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38631141

RESUMO

Pax6 is a critical transcription factor involved in the development of the central nervous system. However, in humans, mutations in Pax6 predominantly result in iris deficiency rather than neurological phenotypes. This may be attributed to the distinct functions of Pax6 isoforms, Pax6a and Pax6b. In this study, we investigated the spatial and temporal expression patterns of Pax6 isoforms during different stages of mouse eye development. We observed a strong correlation between Pax6a expression and the neuroretina gene Sox2, while Pax6b showed a high correlation with iris-component genes, including the mesenchymal gene Foxc1. During early patterning from E10.5, Pax6b was expressed in the hinge of the optic cup and neighboring mesenchymal cells, whereas Pax6a was absent in these regions. At E14.5, both Pax6a and Pax6b were expressed in the future iris and ciliary body, coinciding with the integration of mesenchymal cells and Mitf-positive cells in the outer region. From E18.5, Pax6 isoforms exhibited distinct expression patterns as lineage genes became more restricted. To further validate these findings, we utilized ESC-derived eye organoids, which recapitulated the temporal and spatial expression patterns of lineage genes and Pax6 isoforms. Additionally, we found that the spatial expression patterns of Foxc1 and Mitf were impaired in Pax6b-mutant ESC-derived eye organoids. This in vitro eye organoids model suggested the involvement of Pax6b-positive local mesodermal cells in iris development. These results provide valuable insights into the regulatory roles of Pax6 isoforms during iris and neuroretina development and highlight the potential of ESC-derived eye organoids as a tool for studying normal and pathological eye development.


Assuntos
Olho , Regulação da Expressão Gênica no Desenvolvimento , Organoides , Fator de Transcrição PAX6 , Isoformas de Proteínas , Fator de Transcrição PAX6/genética , Fator de Transcrição PAX6/metabolismo , Animais , Camundongos , Organoides/metabolismo , Organoides/crescimento & desenvolvimento , Organoides/citologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Olho/crescimento & desenvolvimento , Olho/metabolismo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Organogênese/genética
3.
Nature ; 622(7982): 359-366, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37758944

RESUMO

The assembly of cortical circuits involves the generation and migration of interneurons from the ventral to the dorsal forebrain1-3, which has been challenging to study at inaccessible stages of late gestation and early postnatal human development4. Autism spectrum disorder and other neurodevelopmental disorders (NDDs) have been associated with abnormal cortical interneuron development5, but which of these NDD genes affect interneuron generation and migration, and how they mediate these effects remains unknown. We previously developed a platform to study interneuron development and migration in subpallial organoids and forebrain assembloids6. Here we integrate assembloids with CRISPR screening to investigate the involvement of 425 NDD genes in human interneuron development. The first screen aimed at interneuron generation revealed 13 candidate genes, including CSDE1 and SMAD4. We subsequently conducted an interneuron migration screen in more than 1,000 forebrain assembloids that identified 33 candidate genes, including cytoskeleton-related genes and the endoplasmic reticulum-related gene LNPK. We discovered that, during interneuron migration, the endoplasmic reticulum is displaced along the leading neuronal branch before nuclear translocation. LNPK deletion interfered with this endoplasmic reticulum displacement and resulted in abnormal migration. These results highlight the power of this CRISPR-assembloid platform to systematically map NDD genes onto human development and reveal disease mechanisms.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Transtornos do Neurodesenvolvimento , Feminino , Humanos , Recém-Nascido , Gravidez , Movimento Celular/genética , Sistemas CRISPR-Cas/genética , Interneurônios/citologia , Interneurônios/metabolismo , Interneurônios/patologia , Transtornos do Neurodesenvolvimento/genética , Transtornos do Neurodesenvolvimento/patologia , Organoides/citologia , Organoides/embriologia , Organoides/crescimento & desenvolvimento , Organoides/metabolismo , Organoides/patologia , Retículo Endoplasmático/metabolismo , Prosencéfalo/citologia , Prosencéfalo/embriologia , Prosencéfalo/crescimento & desenvolvimento , Prosencéfalo/metabolismo , Prosencéfalo/patologia , Transporte Ativo do Núcleo Celular
4.
Stem Cell Rev Rep ; 19(4): 1116-1123, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36652145

RESUMO

Down syndrome (DS, or trisomy 21, T21), is the most common genetic cause of intellectual disability. Alterations in the complex process of cerebral cortex development contribute to the neurological deficits in DS, although the underlying molecular and cellular mechanisms are not completely understood. Human cerebral organoids (COs) derived from three-dimensional (3D) cultures of induced pluripotent stem cells (iPSCs) provide a new avenue for gaining a better understanding of DS neuropathology. In this study, we aimed to generate iPSCs from individuals with DS (T21-iPSCs) and euploid controls using urine-derived cells, which can be easily and noninvasively obtained from most individuals, and examine their ability to differentiate into neurons and astrocytes grown in monolayer cultures, as well as into 3D COs. We employed nonintegrating episomal vectors to generate urine-derived iPSC lines, and a simple-to-use system to produce COs with forebrain identity. We observed that both T21 and control urine-derived iPSC lines successfully differentiate into neurons and astrocytes in monolayer, as well as into COs that recapitulate early features of human cortical development, including organization of neural progenitor zones, programmed differentiation of excitatory and inhibitory neurons, and upper-and deep-layer cortical neurons as well as astrocytes. Our findings demonstrate for the first time the suitability of using urine-derived iPSC lines to produce COs for modeling DS.


Assuntos
Cérebro , Síndrome de Down , Células-Tronco Pluripotentes Induzidas , Neurogênese , Organoides , Células-Tronco Pluripotentes Induzidas/citologia , Organoides/citologia , Organoides/crescimento & desenvolvimento , Cérebro/citologia , Cérebro/crescimento & desenvolvimento , Síndrome de Down/genética , Síndrome de Down/patologia , Síndrome de Down/urina , Técnicas de Cultura de Células em Três Dimensões , Humanos , Neurônios/citologia , Astrócitos/citologia , Linhagem da Célula
5.
Proc Natl Acad Sci U S A ; 119(30): e2118054119, 2022 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-35858415

RESUMO

Müllerian ducts are paired tubular structures that give rise to most of the female reproductive organs. Any abnormalities in the development and differentiation of these ducts lead to anatomical defects in the female reproductive tract organs categorized as Müllerian duct anomalies. Due to the limited access to fetal tissues, little is understood of human reproductive tract development and the associated anomalies. Although organoids represent a powerful model to decipher human development and disease, such organoids from fetal reproductive organs are not available. Here, we developed organoids from human fetal fallopian tubes and uteri and compared them with their adult counterparts. Our results demonstrate that human fetal reproductive tract epithelia do not express some of the typical markers of adult reproductive tract epithelia. Furthermore, fetal organoids are grossly, histologically, and proteomically different from adult organoids. While external supplementation of WNT ligands or activators in culture medium is an absolute requirement for the adult reproductive tract organoids, fetal organoids are able to grow in WNT-deficient conditions. We also developed decellularized tissue scaffolds from adult human fallopian tubes and uteri. Transplantation of fetal organoids onto these scaffolds led to the regeneration of the adult fallopian tube and uterine epithelia. Importantly, suppression of Wnt signaling, which is altered in patients with Müllerian duct anomalies, inhibits the regenerative ability of human fetal organoids and causes severe anatomical defects in the mouse reproductive tract. Thus, our fetal organoids represent an important platform to study the underlying basis of human female reproductive tract development and diseases.


Assuntos
Tubas Uterinas , Ductos Paramesonéfricos , Organoides , Útero , Adulto , Animais , Tubas Uterinas/crescimento & desenvolvimento , Feminino , Feto , Humanos , Ligantes , Camundongos , Ductos Paramesonéfricos/anormalidades , Organoides/crescimento & desenvolvimento , Organoides/metabolismo , Útero/crescimento & desenvolvimento , Via de Sinalização Wnt
6.
Sci Rep ; 12(1): 7200, 2022 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-35504930

RESUMO

Signaling pathways play an important role in cell fate determination in stem cells and regulate a plethora of developmental programs, the dysregulation of which can lead to human diseases. Growth factors (GFs) regulating these signaling pathways therefore play a major role in the plasticity of adult stem cells and modulate cellular differentiation and tissue repair outcomes. We consider murine mammary organoid generation from self-organizing adult stem cells as a tool to understand the role of GFs in organ development and tissue regeneration. The astounding capacity of mammary organoids to regenerate a gland in vivo after transplantation makes it a convenient model to study organ regeneration. We show organoids grown in suspension with minimal concentration of Matrigel and in the presence of a cocktail of GFs regulating EGF and FGF signaling can recapitulate key epithelial layers of adult mammary gland. We establish a toolkit utilizing in vivo whole animal imaging and ultrasound imaging combined with ex vivo approaches including tissue clearing and confocal imaging to study organ regeneration and ductal morphogenesis. Although the organoid structures were severely impaired in vitro when cultured in the presence of individual GFs, ex vivo imaging revealed ductal branching after transplantation albeit with significantly reduced number of terminal end buds. We anticipate these imaging modalities will open novel avenues to study mammary gland morphogenesis in vivo and can be beneficial for monitoring mammary tumor progression in pre-clinical and clinical settings.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular , Organoides , Animais , Fatores Imunológicos/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Glândulas Mamárias Animais/metabolismo , Camundongos , Morfogênese , Organoides/crescimento & desenvolvimento , Organoides/metabolismo , Regeneração
7.
Cell Rep ; 38(7): 110379, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35172130

RESUMO

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.


Assuntos
Epitélio/crescimento & desenvolvimento , Intestinos/crescimento & desenvolvimento , Organoides/crescimento & desenvolvimento , Membrana Serosa/crescimento & desenvolvimento , Técnicas de Cultura de Tecidos , Alginatos/farmacologia , Adesão Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Colágeno/farmacologia , Combinação de Medicamentos , Epitélio/efeitos dos fármacos , Proteínas Hedgehog/metabolismo , Humanos , Intestinos/ultraestrutura , Laminina/farmacologia , Músculo Liso/citologia , Organoides/efeitos dos fármacos , Organoides/ultraestrutura , Proteoglicanas/farmacologia , Membrana Serosa/efeitos dos fármacos , Membrana Serosa/ultraestrutura , Transdução de Sinais/efeitos dos fármacos , Suspensões , Proteínas Wnt/metabolismo
8.
Life Sci ; 291: 120273, 2022 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-35016877

RESUMO

AIM: Eye organoids are 3D models of the retina that provide new possibilities for studying retinal development, drug toxicity and the molecular mechanisms of diseases. Although there are several protocols that can be used to generate functional tissues, none have been used to assemble human retinal organoids containing mesenchymal stem cells (MSCs). MAIN METHODS: In this study we intend to assess the effective interactions of MSCs and human embryonic stem cells (hESCs) during retinal organoid formation. We evaluated the inducing activities of bone marrow MSCs (BM-MSCs), trabecular meshwork (TM), and stem cells from apical papilla (SCAP)-derived MSCs in differentiation of hESCs in a three-dimensional (3D) direct co-culture system. KEY FINDINGS: In comparison with the two other MSC sources, the induction potential of SCAP was confirmed in the co-culture system. Although the different SCAP cell ratios did not show any significant morphology changes during the first seven days, increasing the number of SCAPs improved formation of the optic vesicle (OV) structure, which was confirmed by assessment of specific markers. The OVs subsequently developed to an optic cup (OC), which was similar to the in vivo environment. These arrangements expressed MITF in the outer layer and CHX10 in the inner layer. SIGNIFICANCE: We assessed the inducing activity of SCAP during differentiation of hESCs towards a retinal fate in a 3D organoid system. However, future studies be conducted to gather additional details about the development of the eye field, retinal differentiation, and the molecular mechanisms of diseases.


Assuntos
Técnicas de Cultura de Células/métodos , Gengiva/citologia , Retina/citologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Olho/citologia , Gengiva/metabolismo , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Organoides/citologia , Organoides/crescimento & desenvolvimento , Organoides/metabolismo , Retina/crescimento & desenvolvimento
9.
Science ; 375(6576): eaaw9021, 2022 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-34990240

RESUMO

Epithelial organoids are stem cell­derived tissues that approximate aspects of real organs, and thus they have potential as powerful tools in basic and translational research. By definition, they self-organize, but the structures formed are often heterogeneous and irreproducible, which limits their use in the lab and clinic. We describe methodologies for spatially and temporally controlling organoid formation, thereby rendering a stochastic process more deterministic. Bioengineered stem cell microenvironments are used to specify the initial geometry of intestinal organoids, which in turn controls their patterning and crypt formation. We leveraged the reproducibility and predictability of the culture to identify the underlying mechanisms of epithelial patterning, which may contribute to reinforcing intestinal regionalization in vivo. By controlling organoid culture, we demonstrate how these structures can be used to answer questions not readily addressable with the standard, more variable, organoid models.


Assuntos
Mucosa Intestinal/crescimento & desenvolvimento , Organogênese , Organoides/crescimento & desenvolvimento , Engenharia Tecidual , Animais , Diferenciação Celular , Forma Celular , Células Epiteliais/citologia , Hidrogéis , Mucosa Intestinal/anatomia & histologia , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Camundongos , Organoides/anatomia & histologia , Organoides/citologia , Organoides/metabolismo , Celulas de Paneth/citologia , Receptores Notch/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/fisiologia , Técnicas de Cultura de Tecidos , Proteínas de Sinalização YAP/metabolismo
10.
PLoS One ; 17(1): e0262950, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35073389

RESUMO

This study presents novel biocompatible Polydimethylsiloxane (PDMS)-based micromechanical tweezers (µTweezers) capable of the stiffness characterization and manipulation of hydrogel-based organoids. The system showed great potential for complementing established mechanical characterization methods such as Atomic Force Microscopy (AFM), parallel plate compression (PPC), and nanoindentation, while significantly reducing the volume of valuable hydrogels used for testing. We achieved a volume reduction of ~0.22 µl/sample using the µTweezers vs. ~157 µl/sample using the PPC, while targeting high-throughput measurement of widely adopted micro-mesoscale (a few hundred µm-1500 µm) 3D cell cultures. The µTweezers applied and measured nano-millinewton forces through cantilever' deflection with high linearity and tunability for different applications; the assembly is compatible with typical inverted optical microscopes and fit on standard tissue culture Petri dishes, allowing mechanical compression characterization of arrayed 3D hydrogel-based organoids in a high throughput manner. The average achievable output per group was 40 tests per hour, where 20 organoids and 20 reference images in one 35 mm petri dish were tested, illustrating efficient productivity to match the increasing demand on 3D organoids' applications. The changes in stiffness of collagen I hydrogel organoids in four conditions were measured, with ovarian cancer cells (SKOV3) or without (control). The Young's modulus of the control group (Control-day 0, E = 407± 146, n = 4) measured by PPC was used as a reference modulus, where the relative elastic compressive modulus of the other groups based on the stiffness measurements was also calculated (control-day 0, E = 407 Pa), (SKOV3-day 0, E = 318 Pa), (control-day 5, E = 528 Pa), and (SKOV3-day 5, E = 376 Pa). The SKOV3-embedded hydrogel-based organoids had more shrinkage and lowered moduli on day 0 and day 5 than controls, consistently, while SKOV3 embedded organoids increased in stiffness in a similar trend to the collagen I control from day 0 to day 5. The proposed method can contribute to the biomedical, biochemical, and regenerative engineering fields, where bulk mechanical characterization is of interest. The µTweezers will also provide attractive design and application concepts to soft membrane-micro 3D robotics, sensors, and actuators.


Assuntos
Técnicas de Cultura de Células , Hidrogéis/química , Organoides , Estresse Mecânico , Linhagem Celular Tumoral , Feminino , Humanos , Pessoa de Meia-Idade , Organoides/citologia , Organoides/crescimento & desenvolvimento
11.
Cell Immunol ; 371: 104458, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34847407

RESUMO

Our previous work suggested that high SIRT1 expression by cancer cells predicted a poor colorectal cancer (CRC) prognosis, but its role in the tumor microenvironment was unclear. Here, we examined tumor-infiltrating lymphocytes (TILs) in CRC expressing different levels of SIRT1. We also established a co-culture system with monocytes, CD8+ T cells and patient-derived tumor organoids (PDOs) to study the relationships between immune cells and cancer cells. The percentage of CD8+ T cells was decreased and the percentage of macrophages was increased in SIRT1-high (SIRT1-hi) CRC. Co-culture results showed that tumor-associated macrophages (TAMs) from SIRT1-hi CRC inhibited the proliferation and anti-tumor activity of CD8+ T cells. Importantly, SIRT1-hi CRC were shown to modulate the migration and the activity of TAMs. RNA sequencing revealed that CD14+ monocytes in SIRT1-hi patients expressed higher levels of CXCR4. Mechanistically, SIRT1 expression was shown to promote CXCL12 expression by inhibiting the acetylation of p53. Our findings indicate that SIRT1 in CRC induces TAM migration through the CXCR4/CXCL12 pathway, and inhibits the proliferation and activity of CD8+ T cells, resulting in promotion of CRC progression.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Quimiocina CXCL12/metabolismo , Neoplasias Colorretais/imunologia , Macrófagos/imunologia , Receptores CXCR4/metabolismo , Sirtuína 1/metabolismo , Diferenciação Celular/imunologia , Linhagem Celular Tumoral , Movimento Celular/imunologia , Técnicas de Cocultura , Neoplasias Colorretais/patologia , Células HT29 , Humanos , Linfócitos do Interstício Tumoral/imunologia , Organoides/crescimento & desenvolvimento , Interferência de RNA , RNA Interferente Pequeno/genética , Sirtuína 1/genética , Microambiente Tumoral/imunologia
12.
Gene ; 813: 146131, 2022 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-34933077

RESUMO

The retina is a complex system containing several neuron types arranged in distinct layers. Many aspects of the retina's development and the molecular events in the human light-sensing system have been previously unveiled. However, there is limited information about regulatory networks governing the transitional stages during retina development. To address this issue, we have studied the transcriptome dynamics of mice-derived retinal organoid development in 10 successive time-points, from stem cell to functional retina. For the first time, we have identified the main modules of genes related to different stages of development and predicted all possible transcription factors. A major shift in the transcriptome occurs during the transition of cells from D0 to D10 and again at the late stages of retina development. Transcription, nervous system development, cell cycle, neurotransmitter transport, glycosylation, and lipid metabolisms are the most important biological processes during retina development. Altogether, we have identified and reported 15 TFs, including Irx2, Irx3, Lmo2, Tead2, Tbx20, and Zeb1, which are potentially involved in the regulation of retinal organoid development. In conclusion, using several rigorous analyses, we have found main stage-specific biological processes in the retina development and predicted TFs with strong potency in controlling this structure.


Assuntos
Redes Reguladoras de Genes , Organoides/fisiologia , Retina/fisiologia , Animais , Diferenciação Celular/genética , Bases de Dados Genéticas , Expressão Gênica , Proteínas de Homeodomínio/genética , Camundongos , Organogênese/genética , Organoides/crescimento & desenvolvimento , Organoides/metabolismo , Retina/crescimento & desenvolvimento , Retina/metabolismo , Proteínas com Domínio T/genética , Fatores de Transcrição/genética , Transcriptoma
13.
Cell ; 185(1): 42-61, 2022 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-34774127

RESUMO

The construction of the human nervous system is a distinctly complex although highly regulated process. Human tissue inaccessibility has impeded a molecular understanding of the developmental specializations from which our unique cognitive capacities arise. A confluence of recent technological advances in genomics and stem cell-based tissue modeling is laying the foundation for a new understanding of human neural development and dysfunction in neuropsychiatric disease. Here, we review recent progress on uncovering the cellular and molecular principles of human brain organogenesis in vivo as well as using organoids and assembloids in vitro to model features of human evolution and disease.


Assuntos
Transtorno do Espectro Autista/metabolismo , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Epilepsia/metabolismo , Neurogênese/fisiologia , Esquizofrenia/metabolismo , Animais , Transtorno do Espectro Autista/genética , Encéfalo/metabolismo , Epilepsia/genética , Humanos , Mutação , Neurônios/citologia , Neurônios/metabolismo , Organoides/embriologia , Organoides/crescimento & desenvolvimento , Esquizofrenia/genética
14.
Nat Commun ; 12(1): 7302, 2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34911939

RESUMO

Three-dimensional brain organoids have emerged as a valuable model system for studies of human brain development and pathology. Here we establish a midbrain organoid culture system to study the developmental trajectory from pluripotent stem cells to mature dopamine neurons. Using single cell RNA sequencing, we identify the presence of three molecularly distinct subtypes of human dopamine neurons with high similarity to those in developing and adult human midbrain. However, despite significant advancements in the field, the use of brain organoids can be limited by issues of reproducibility and incomplete maturation which was also observed in this study. We therefore designed bioengineered ventral midbrain organoids supported by recombinant spider-silk microfibers functionalized with full-length human laminin. We show that silk organoids reproduce key molecular aspects of dopamine neurogenesis and reduce inter-organoid variability in terms of cell type composition and dopamine neuron formation.


Assuntos
Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Dopamina/metabolismo , Neurônios/metabolismo , Organoides/crescimento & desenvolvimento , Encéfalo/citologia , Humanos , Neurogênese , Neurônios/citologia , Organoides/citologia , Organoides/metabolismo , Análise de Sequência de RNA , Análise de Célula Única , Transcriptoma
15.
Development ; 148(21)2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34751748

RESUMO

Although the role of the transcription factor NF-κB in intestinal inflammation and tumor formation has been investigated extensively, a physiological function of NF-κB in sustaining intestinal epithelial homeostasis beyond inflammation has not been demonstrated. Using NF-κB reporter mice, we detected strong NF-κB activity in Paneth cells, in '+4/+5' secretory progenitors and in scattered Lgr5+ crypt base columnar stem cells of small intestinal (SI) crypts. To examine NF-κB functions in SI epithelial self-renewal, mice or SI crypt organoids ('mini-guts') with ubiquitously suppressed NF-κB activity were used. We show that NF-κB activity is dispensable for maintaining SI epithelial proliferation, but is essential for ex vivo organoid growth. Furthermore, we demonstrate a dramatic reduction of Paneth cells in the absence of NF-κB activity, concomitant with a significant increase in goblet cells and immature intermediate cells. This indicates that NF-κB is required for proper Paneth versus goblet cell differentiation and for SI epithelial homeostasis, which occurs via regulation of Wnt signaling and Sox9 expression downstream of NF-κB. The current study thus presents evidence for an important role for NF-κB in intestinal epithelial self-renewal.


Assuntos
Células Caliciformes/citologia , Intestino Delgado/citologia , NF-kappa B/metabolismo , Celulas de Paneth/citologia , Animais , Diferenciação Celular , Autorrenovação Celular , Células Caliciformes/metabolismo , Homeostase , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Intestino Delgado/patologia , Camundongos , NF-kappa B/genética , Organoides/citologia , Organoides/crescimento & desenvolvimento , Organoides/metabolismo , Celulas de Paneth/metabolismo , Fatores de Transcrição SOX9/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt
16.
Development ; 148(21)2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34651188

RESUMO

Two recently developed models, trophoblast organoids and trophoblast stem cells (TSCs), are useful tools to further the understanding of human placental development. Both differentiate from villous cytotrophoblast (VCT) to either extravillous trophoblast (EVT) or syncytiotrophoblast (SCT). Here, we compare the transcriptomes and miRNA profiles of these models to identify which trophoblast they resemble in vivo. Our findings indicate that TSCs do not readily undergo SCT differentiation and closely resemble cells at the base of the cell columns from where EVT derives. In contrast, organoids are similar to VCT and undergo spontaneous SCT differentiation. A defining feature of human trophoblast is that VCT and SCT are human leukocyte antigen (HLA) null, whereas EVT expresses HLA-C, -G and -E molecules. We find that trophoblast organoids retain these in vivo characteristics. In contrast, TSCs express classical HLA-A and HLA-B molecules, and maintain their expression after EVT differentiation, with upregulation of HLA-G. Furthermore, HLA expression in TSCs differs when grown in 3D rather than in 2D, suggesting that mechanical cues are important. Our results can be used to select the most suitable model for the study of trophoblast development, function and pathology.


Assuntos
Modelos Biológicos , Trofoblastos/citologia , Técnicas de Cultura de Células , Diferenciação Celular , Células Cultivadas , Feminino , Antígenos HLA/genética , Antígenos HLA/metabolismo , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Organoides/citologia , Organoides/crescimento & desenvolvimento , Organoides/metabolismo , Placentação , Gravidez , Células-Tronco/citologia , Células-Tronco/metabolismo , Transcriptoma , Trofoblastos/metabolismo
17.
Adv Mater ; 33(45): e2007949, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34561899

RESUMO

Recent advances in 3D cell culture technology have enabled scientists to generate stem cell derived organoids that recapitulate the structural and functional characteristics of native organs. Current organoid technologies have been striding toward identifying the essential factors for controlling the processes involved in organoid development, including physical cues and biochemical signaling. There is a growing demand for engineering dynamic niches characterized by conditions that resemble in vivo organogenesis to generate reproducible and reliable organoids for various applications. Innovative biomaterial-based and advanced engineering-based approaches have been incorporated into conventional organoid culture methods to facilitate the development of organoid research. The recent advances in organoid engineering, including extracellular matrices and genetic modulation, are comprehensively summarized to pinpoint the parameters critical for organ-specific patterning. Moreover, perspective trends in developing tunable organoids in response to exogenous and endogenous cues are discussed for next-generation developmental studies, disease modeling, and therapeutics.


Assuntos
Engenharia Biomédica , Técnicas de Cultura de Células em Três Dimensões/métodos , Organoides/metabolismo , Materiais Biocompatíveis/química , Matriz Extracelular/metabolismo , Engenharia Genética , Humanos , Hidrogéis/química , Neoplasias/genética , Neoplasias/patologia , Organoides/citologia , Organoides/crescimento & desenvolvimento , Células-Tronco/citologia , Células-Tronco/metabolismo
19.
Int J Mol Sci ; 22(16)2021 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-34445064

RESUMO

Photoreceptors are critical components of the retina and play a role in the first step of the conversion of light to electric signals. With the discovery of the intrinsically photosensitive retinal ganglion cells, which regulate non-image-forming visual processes, our knowledge of the photosensitive cell family in the retina has deepened. Photoreceptor development is regulated by specific genes and proteins and involves a series of molecular processes including DNA transcription, post-transcriptional modification, protein translation, and post-translational modification. Single-cell sequencing is a promising technology for the study of photoreceptor development. This review presents an overview of the types of human photoreceptors, summarizes recent discoveries in the regulatory mechanisms underlying their development at single-cell resolution, and outlines the prospects in this field.


Assuntos
Células Fotorreceptoras de Vertebrados/citologia , Retina/crescimento & desenvolvimento , Análise de Célula Única/métodos , Animais , Humanos , Organoides/citologia , Organoides/embriologia , Organoides/crescimento & desenvolvimento , Células Fotorreceptoras de Vertebrados/metabolismo , Retina/citologia , Retina/embriologia
20.
Nat Commun ; 12(1): 4730, 2021 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-34354063

RESUMO

Brain organoids derived from human pluripotent stem cells provide a highly valuable in vitro model to recapitulate human brain development and neurological diseases. However, the current systems for brain organoid culture require further improvement for the reliable production of high-quality organoids. Here, we demonstrate two engineering elements to improve human brain organoid culture, (1) a human brain extracellular matrix to provide brain-specific cues and (2) a microfluidic device with periodic flow to improve the survival and reduce the variability of organoids. A three-dimensional culture modified with brain extracellular matrix significantly enhanced neurogenesis in developing brain organoids from human induced pluripotent stem cells. Cortical layer development, volumetric augmentation, and electrophysiological function of human brain organoids were further improved in a reproducible manner by dynamic culture in microfluidic chamber devices. Our engineering concept of reconstituting brain-mimetic microenvironments facilitates the development of a reliable culture platform for brain organoids, enabling effective modeling and drug development for human brain diseases.


Assuntos
Encéfalo/crescimento & desenvolvimento , Encéfalo/fisiologia , Dispositivos Lab-On-A-Chip , Neurogênese/fisiologia , Organoides/crescimento & desenvolvimento , Organoides/fisiologia , Animais , Encéfalo/citologia , Meios de Cultura , Fenômenos Eletrofisiológicos , Matriz Extracelular/fisiologia , Estudos de Viabilidade , Perfilação da Expressão Gênica , Humanos , Hidrogéis , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Modelos Anatômicos , Modelos Neurológicos , Neurogênese/genética , Neuroglia/citologia , Neuroglia/fisiologia , Técnicas de Cultura de Órgãos/instrumentação , Técnicas de Cultura de Órgãos/métodos , Organoides/citologia , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA