Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 4976, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38862520

RESUMO

Twisted gastrulation (TWSG1) is an evolutionarily conserved secreted glycoprotein which controls signaling by Bone Morphogenetic Proteins (BMPs). TWSG1 binds BMPs and their antagonist Chordin to control BMP signaling during embryonic development, kidney regeneration and cancer. We report crystal structures of TWSG1 alone and in complex with a BMP ligand, Growth Differentiation Factor 5. TWSG1 is composed of two distinct, disulfide-rich domains. The TWSG1 N-terminal domain occupies the BMP type 1 receptor binding site on BMPs, whereas the C-terminal domain binds to a Chordin family member. We show that TWSG1 inhibits BMP function in cellular signaling assays and mouse colon organoids. This inhibitory function is abolished in a TWSG1 mutant that cannot bind BMPs. The same mutation in the Drosophila TWSG1 ortholog Tsg fails to mediate BMP gradient formation required for dorsal-ventral axis patterning of the early embryo. Our studies reveal the evolutionarily conserved mechanism of BMP signaling inhibition by TWSG1.


Assuntos
Proteínas Morfogenéticas Ósseas , Transdução de Sinais , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Morfogenéticas Ósseas/genética , Camundongos , Humanos , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/química , Glicoproteínas/metabolismo , Glicoproteínas/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Sítios de Ligação , Domínios Proteicos , Ligação Proteica , Organoides/metabolismo , Organoides/embriologia , Células HEK293 , Gastrulação/genética , Mutação , Cristalografia por Raios X , Drosophila melanogaster/embriologia , Drosophila melanogaster/metabolismo , Drosophila melanogaster/genética , Proteínas
2.
Nature ; 620(7974): 615-624, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37558872

RESUMO

The concomitant occurrence of tissue growth and organization is a hallmark of organismal development1-3. This often means that proliferating and differentiating cells are found at the same time in a continuously changing tissue environment. How cells adapt to architectural changes to prevent spatial interference remains unclear. Here, to understand how cell movements that are key for growth and organization are orchestrated, we study the emergence of photoreceptor neurons that occur during the peak of retinal growth, using zebrafish, human tissue and human organoids. Quantitative imaging reveals that successful retinal morphogenesis depends on the active bidirectional translocation of photoreceptors, leading to a transient transfer of the entire cell population away from the apical proliferative zone. This pattern of migration is driven by cytoskeletal machineries that differ depending on the direction: microtubules are exclusively required for basal translocation, whereas actomyosin is involved in apical movement. Blocking the basal translocation of photoreceptors induces apical congestion, which hampers the apical divisions of progenitor cells and leads to secondary defects in lamination. Thus, photoreceptor migration is crucial to prevent competition for space, and to allow concurrent tissue growth and lamination. This shows that neuronal migration, in addition to its canonical role in cell positioning4, can be involved in coordinating morphogenesis.


Assuntos
Movimento Celular , Morfogênese , Células Fotorreceptoras , Retina , Animais , Humanos , Actomiosina/metabolismo , Competição entre as Células , Diferenciação Celular , Movimento Celular/fisiologia , Proliferação de Células , Microtúbulos/metabolismo , Morfogênese/fisiologia , Organoides/citologia , Organoides/embriologia , Células Fotorreceptoras/citologia , Células Fotorreceptoras/fisiologia , Retina/citologia , Retina/embriologia , Peixe-Zebra/embriologia
3.
Cell ; 184(12): 3299-3317.e22, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-34019794

RESUMO

Organoids capable of forming tissue-like structures have transformed our ability to model human development and disease. With the notable exception of the human heart, lineage-specific self-organizing organoids have been reported for all major organs. Here, we established self-organizing cardioids from human pluripotent stem cells that intrinsically specify, pattern, and morph into chamber-like structures containing a cavity. Cardioid complexity can be controlled by signaling that instructs the separation of cardiomyocyte and endothelial layers and by directing epicardial spreading, inward migration, and differentiation. We find that cavity morphogenesis is governed by a mesodermal WNT-BMP signaling axis and requires its target HAND1, a transcription factor linked to developmental heart chamber defects. Upon cryoinjury, cardioids initiated a cell-type-dependent accumulation of extracellular matrix, an early hallmark of both regeneration and heart disease. Thus, human cardioids represent a powerful platform to mechanistically dissect self-organization, congenital heart defects and serve as a foundation for future translational research.


Assuntos
Coração/embriologia , Organogênese , Organoides/embriologia , Ativinas/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Cálcio/metabolismo , Linhagem Celular , Linhagem da Célula , Galinhas , Células Endoteliais/citologia , Proteínas da Matriz Extracelular/metabolismo , Feminino , Fibroblastos/citologia , Proteína Homeobox Nkx-2.5/metabolismo , Humanos , Masculino , Mesoderma/embriologia , Modelos Biológicos , Miocárdio/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteínas Wnt/metabolismo
4.
Cell ; 184(12): 3281-3298.e22, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-34019796

RESUMO

Organs are composed of diverse cell types that traverse transient states during organogenesis. To interrogate this diversity during human development, we generate a single-cell transcriptome atlas from multiple developing endodermal organs of the respiratory and gastrointestinal tract. We illuminate cell states, transcription factors, and organ-specific epithelial stem cell and mesenchyme interactions across lineages. We implement the atlas as a high-dimensional search space to benchmark human pluripotent stem cell (hPSC)-derived intestinal organoids (HIOs) under multiple culture conditions. We show that HIOs recapitulate reference cell states and use HIOs to reconstruct the molecular dynamics of intestinal epithelium and mesenchyme emergence. We show that the mesenchyme-derived niche cue NRG1 enhances intestinal stem cell maturation in vitro and that the homeobox transcription factor CDX2 is required for regionalization of intestinal epithelium and mesenchyme in humans. This work combines cell atlases and organoid technologies to understand how human organ development is orchestrated.


Assuntos
Anatomia Artística , Atlas como Assunto , Desenvolvimento Embrionário , Endoderma/embriologia , Modelos Biológicos , Organoides/embriologia , Fator de Transcrição CDX2/metabolismo , Linhagem Celular , Fator de Crescimento Epidérmico/farmacologia , Células Epiteliais/citologia , Feminino , Gastrulação , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Intestinos/embriologia , Masculino , Mesoderma/embriologia , Pessoa de Meia-Idade , Neuregulina-1/metabolismo , Especificidade de Órgãos , Células-Tronco Pluripotentes/citologia
5.
Stem Cell Reports ; 16(2): 354-369, 2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33482102

RESUMO

Floating spheroidal aggregates of mouse embryonic stem cells can develop into polarized/elongated organoids, namely gastruloids. We set up a high-performing assay to measure gastruloid formation efficiency (GFE), and found that GFE decreases as pluripotency progresses from naive (GFE ≥ 95%) to primed (GFE = 0) state. Specifically, we show that primed EpiSCs fail to generate proper cell aggregates, while early-primed EpiLCs aggregate but eventually fail to develop into elongated gastruloids. Moreover, we characterized proline-induced cells (PiCs), a LIF-dependent reversible early-primed state of pluripotency, and show that PiCs are able to generate gastruloids (GFE ∼ 50%) and are also competent to differentiate into primordial germ cell-like cells. Thus, we propose the GFE assay as a valuable functional tool to discriminate different states of the pluripotency continuum.


Assuntos
Desenvolvimento Embrionário , Fator de Crescimento Epidérmico/metabolismo , Camadas Germinativas/metabolismo , Glicoproteínas de Membrana/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Proteínas de Neoplasias/metabolismo , Organogênese , Organoides/embriologia , Células-Tronco Pluripotentes/metabolismo , Animais , Diferenciação Celular , Fator de Crescimento Epidérmico/genética , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/genética
6.
Nature ; 585(7826): 574-578, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32939089

RESUMO

Epithelial organoids, such as those derived from stem cells of the intestine, have great potential for modelling tissue and disease biology1-4. However, the approaches that are used at present to derive these organoids in three-dimensional matrices5,6 result in stochastically developing tissues with a closed, cystic architecture that restricts lifespan and size, limits experimental manipulation and prohibits homeostasis. Here, by using tissue engineering and the intrinsic self-organization properties of cells, we induce intestinal stem cells to form tube-shaped epithelia with an accessible lumen and a similar spatial arrangement of crypt- and villus-like domains to that in vivo. When connected to an external pumping system, the mini-gut tubes are perfusable; this allows the continuous removal of dead cells to prolong tissue lifespan by several weeks, and also enables the tubes to be colonized with microorganisms for modelling host-microorganism interactions. The mini-intestines include rare, specialized cell types that are seldom found in conventional organoids. They retain key physiological hallmarks of the intestine and have a notable capacity to regenerate. Our concept for extrinsically guiding the self-organization of stem cells into functional organoids-on-a-chip is broadly applicable and will enable the attainment of more physiologically relevant organoid shapes, sizes and functions.


Assuntos
Homeostase , Intestinos/embriologia , Morfogênese , Organoides/embriologia , Alicerces Teciduais , Animais , Padronização Corporal , Diferenciação Celular , Linhagem da Célula , Cryptosporidium parvum/patogenicidade , Células-Tronco Embrionárias Humanas/citologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Intestinos/citologia , Intestinos/parasitologia , Intestinos/patologia , Camundongos , Modelos Biológicos , Organoides/citologia , Organoides/parasitologia , Organoides/patologia , Regeneração , Medicina Regenerativa , Células-Tronco , Técnicas de Cultura de Tecidos/métodos , Engenharia Tecidual
7.
Mol Biol Rep ; 47(9): 6621-6633, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32803508

RESUMO

Purple coneflower (Echinacea purpurea (L.) Moench) is a widely used medicinal and ornamental plant. In the present study, the callus embryogenesis was examined using benzyl adenine (BA) at three levels (3, 4, 5 mg L-1), 1-Naphthalene acetic acid (NAA) at three levels (0.1, 0.2 and 0.5 mg L-1) with or without activated charcoal (1 g L-1), coconut milk (50 ml L-1) and casein hydrolysate (50 mg L-1) in the MS (Murashige and Skoog 1962) medium. The embryogenesis indirectly occurred with the production of callus. The calli were observed in three forms: undifferentiated, embryogenic and organogenic. The embryogenic calli were dark green and coherent with a faster growth rate. The highest embryogenesis (100%) and embryonic regeneration (plantlet production) were obtained in the combined BA + NAA treatments with the activated charcoal, coconut milk and casein hydrolysate. However, the combined treatments of growth regulators failed to produce somatic embryos without the use of coconut milk and casein hydrolysate. The maximum amount of protein, peroxidase and catalase activity of embryogenic calli (2.02, 1.79 and 6.62ΔOD/Min/mg.protein, respectively), and highest percentage of acclimatization success (29.3% of plants) were obtained in the combined treatment of 5 mg L-1 BA + 0.5 mg L-1 NAA + activated charcoal + coconut milk + casein hydrolysate. The highest amount of chlorophyll content (33.3 SPAD value) and growth characteristics of acclimatized plantlets were observed in the media containing 3 mg L-1 BA + 0.1 and 0.2 mg L-1 NAA + 1 g. L-1 combined activated charcoal, coconut milk, casein hydrolysate. The histological studies confirmed the somatic embryogenesis in purple coneflower. Generally, it was found that the somatic embryogenesis of E. purpurea occurs at high levels of BA and low levels of NAA with the addition of coconut milk and casein hydrolysate.


Assuntos
Antioxidantes/farmacologia , Echinacea/química , Echinacea/embriologia , Reguladores de Crescimento de Plantas/farmacologia , Brotos de Planta/efeitos dos fármacos , Técnicas de Embriogênese Somática de Plantas/métodos , Adenina/análogos & derivados , Adenina/farmacologia , Caseínas/farmacologia , Carvão Vegetal/farmacologia , Cocos/química , Meios de Cultura , Echinacea/enzimologia , Ácidos Naftalenoacéticos/farmacologia , Organoides/citologia , Organoides/efeitos dos fármacos , Organoides/embriologia , Organoides/crescimento & desenvolvimento , Brotos de Planta/embriologia , Brotos de Planta/crescimento & desenvolvimento , Plantas Medicinais/química
8.
J Clin Invest ; 130(8): 4396-4410, 2020 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-32427591

RESUMO

Esophageal atresia (EA/TEF) is a common congenital abnormality present in 1 of 4000 births. Here we show that atretic esophagi lack Noggin (NOG) expression, resulting in immature esophagus that contains respiratory glands. Moreover, when using mouse esophageal organoid units (EOUs) or tracheal organoid units (TOUs) as a model of foregut development and differentiation in vitro, NOG determines whether foregut progenitors differentiate toward esophageal or tracheal epithelium. These results indicate that NOG is a critical regulator of cell fate decisions between esophageal and pulmonary morphogenesis, and its lack of expression results in EA/TEF.


Assuntos
Proteínas de Transporte/metabolismo , Diferenciação Celular , Atresia Esofágica/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Modelos Biológicos , Células-Tronco/metabolismo , Animais , Proteínas de Transporte/genética , Linhagem Celular , Atresia Esofágica/genética , Atresia Esofágica/patologia , Humanos , Camundongos , Organoides/embriologia , Organoides/patologia , Células-Tronco/patologia
9.
Dev Biol ; 463(2): 101-109, 2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32422143

RESUMO

Loss of expression of the transcription regulator DC-SCRIPT (Zfp366) is a prominent prognostic event in estrogen receptor-positive breast cancer patients. Studying the inherent link between breast morphogenesis and tumorigenesis, we recently reported that DC-SCRIPT affects normal mammary branching morphogenesis and mammary epithelium homeostasis. Here we investigated the molecular mechanism involved in DC-SCRIPT mediated regulation of FGF2 induced mammary branching morphogenesis in a 3D organoid culture system. Our data show that the delayed mammary organoid branching observed in DC-SCRIPT-/- organoids cannot be compensated for by increasing FGF2 levels. Interestingly, FGFR1, the dominant FGF2 receptor, was expressed at a significantly lower level in basal epithelial cells of DC-SCRIPT deficient organoids relative to wildtype organoids. A potential link between DC-SCRIPT and FGFR1 was further supported by the predicted locations of the DC-SCRIPT DNA binding motif at the Fgfr1 gene. Moreover, ERK1/2 phosphorylation downstream of the FGFR1 pathway was decreased in basal epithelial cells of DC-SCRIPT deficient organoids. Altogether, this study shows a relationship between DC-SCRIPT and FGFR1 related pERK signaling in modulating the branching morphogenesis of mammary organoids in vitro.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Glândulas Mamárias Animais/embriologia , Proteínas Nucleares/metabolismo , Organogênese , Organoides/embriologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Fatores de Transcrição/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Feminino , Sistema de Sinalização das MAP Quinases , Glândulas Mamárias Animais/citologia , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Organoides/citologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Fatores de Transcrição/genética
10.
J Vis Exp ; (153)2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31789309

RESUMO

At the end of the suckling period, many mammalian species undergo major changes in the intestinal epithelium that are associated with the capability to digest solid food. This process is termed suckling-to-weaning transition and results in the replacement of neonatal epithelium with adult epithelium which goes hand in hand with metabolic and morphological adjustments. These complex developmental changes are the result of a genetic program that is intrinsic to the intestinal epithelial cells but can, to some extent, be modulated by extrinsic factors. Prolonged culture of mouse primary intestinal epithelial cells from late fetal period, recapitulates suckling-to-weaning transition in vitro. Here, we describe a detailed protocol for mouse fetal intestinal organoid culture best suited to model this process in vitro. We describe several useful assays designed to monitor the change of intestinal functions associated with suckling-to-weaning transition over time. Additionally, we include an example of an extrinsic factor that is capable to affect suckling-to-weaning transition in vivo, as a representation of modulating the timing of suckling-to-weaning transition in vitro. This in vitro approach can be used to study molecular mechanisms of the suckling-to-weaning transition as well as modulators of this process. Importantly, with respect to animal ethics in research, replacing in vivo models by this in vitro model contributes to refinement of animal experiments and possibly to a reduction in the use of animals to study gut maturation processes.


Assuntos
Animais Lactentes/fisiologia , Desenvolvimento Fetal/fisiologia , Mucosa Intestinal/embriologia , Mucosa Intestinal/fisiologia , Organoides/embriologia , Organoides/fisiologia , Animais , Células Cultivadas , Células Epiteliais/fisiologia , Mucosa Intestinal/citologia , Camundongos , Técnicas de Cultura de Órgãos , Organoides/citologia , Desmame
11.
Semin Cell Dev Biol ; 95: 93-97, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30904636

RESUMO

The ability to reproduce early stages of human neurodevelopment in the laboratory is one of the most exciting fields in modern neuroscience. The inaccessibility of the healthy human brain developing in utero has delayed our understanding of the initial steps in the formation of one of the most complex tissues in the body. Animal models, postmortem human tissues and cellular systems have been instrumental in contributing to our understanding of the human brain. However, all model systems have intrinsic limitations. The emerging field of brain organoids, which are three-dimensional self-assembled multicellular structures derived from human pluripotent stem cells, offers a promising complementary cellular model for the study of the human brain. Here, we will discuss the initial experiments that were the foundation for this emerging field, highlight recent uses of the technology and offer our perspective on future directions that might guide further exploratory experimentation to improve the human brain organoid model system.


Assuntos
Encéfalo/embriologia , Modelos Biológicos , Doenças do Sistema Nervoso/patologia , Organoides/embriologia , Encéfalo/efeitos dos fármacos , Encéfalo/virologia , Poluentes Ambientais/toxicidade , Humanos , Doenças do Sistema Nervoso/genética , Organoides/efeitos dos fármacos , Vírus/metabolismo
12.
Cell Rep ; 22(1): 242-254, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29298425

RESUMO

The mammalian hair follicle arises during embryonic development from coordinated interactions between the epidermis and dermis. It is currently unclear how to recapitulate hair follicle induction in pluripotent stem cell cultures for use in basic research studies or in vitro drug testing. To date, generation of hair follicles in vitro has only been possible using primary cells isolated from embryonic skin, cultured alone or in a co-culture with stem cell-derived cells, combined with in vivo transplantation. Here, we describe the derivation of skin organoids, constituting epidermal and dermal layers, from a homogeneous population of mouse pluripotent stem cells in a 3D culture. We show that skin organoids spontaneously produce de novo hair follicles in a process that mimics normal embryonic hair folliculogenesis. This in vitro model of skin development will be useful for studying mechanisms of hair follicle induction, evaluating hair growth or inhibitory drugs, and modeling skin diseases.


Assuntos
Embrião de Mamíferos , Folículo Piloso , Células-Tronco Pluripotentes Induzidas , Organoides , Animais , Técnicas de Cultura de Células , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Folículo Piloso/citologia , Folículo Piloso/embriologia , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Organoides/citologia , Organoides/embriologia
13.
Cell Rep ; 21(10): 2661-2670, 2017 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-29212014

RESUMO

Organoid technology provides a revolutionary paradigm toward therapy but has yet to be applied in humans, mainly because of reproducibility and scalability challenges. Here, we overcome these limitations by evolving a scalable organ bud production platform entirely from human induced pluripotent stem cells (iPSC). By conducting massive "reverse" screen experiments, we identified three progenitor populations that can effectively generate liver buds in a highly reproducible manner: hepatic endoderm, endothelium, and septum mesenchyme. Furthermore, we achieved human scalability by developing an omni-well-array culture platform for mass producing homogeneous and miniaturized liver buds on a clinically relevant large scale (>108). Vascularized and functional liver tissues generated entirely from iPSCs significantly improved subsequent hepatic functionalization potentiated by stage-matched developmental progenitor interactions, enabling functional rescue against acute liver failure via transplantation. Overall, our study provides a stringent manufacturing platform for multicellular organoid supply, thus facilitating clinical and pharmaceutical applications especially for the treatment of liver diseases through multi-industrial collaborations.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Fígado/enzimologia , Organoides/citologia , Organoides/embriologia , Células-Tronco Pluripotentes/citologia , Diferenciação Celular/fisiologia , Células Cultivadas , Humanos , Fígado/citologia
14.
Artigo em Inglês | MEDLINE | ID: mdl-28750869

RESUMO

The intestinal epithelial cells reside in close proximity to myofibroblasts and microbiota, which are supposed to have an impact on intestinal stem cells fate and to influence processes of tissue maturation and regeneration. Mechanism underlying these phenomena and their diversity among vertebrates can be studied in 3D organoid cultures. We investigated the growth of chicken embryo intestinal epithelial organoids in Matrigel with and without Toll-like receptors (TLRs) stimulation. The organoid cultures contained also some myofibroblasts with potential to promote intestinal stem cell survival. Organoid cells, expressing TLR4, TLR2 type 1 and TLR2 type 2 were incubated with their agonists (lipopolysaccharide - LPS and Pam3CSK4) or co-cultured with Lactobacillus acidophilus bacteria (LA-5). Pam3CSK4 and LA-5 promoted organoid growth, which was demonstrated by comparing the morphological parameters (mean number and area of organoids). The profile of prostaglandins (PG), known to promote intestinal regeneration, in supernatants from organoid and fibroblast cultures were evaluated. Both PGE2 and PGD2 were detected. As compared to unstimulated controls, supernatants from the Pam3CSK4-stimulated organoids contained twice as much of PGE2 and PGD2. The changes in production of prostaglandins and the support of epithelial cell growth by myofibroblasts are factors potentially responsible for stimulatory effect of TLR2 activation.


Assuntos
Mucosa Intestinal/embriologia , Lactobacillus acidophilus/fisiologia , Lipopeptídeos/farmacologia , Organoides/embriologia , Probióticos , Receptor 2 Toll-Like/agonistas , Receptor 2 Toll-Like/imunologia , Animais , Embrião de Galinha , Técnicas de Cocultura , Dinoprostona/biossíntese , Células Epiteliais/fisiologia , Mucosa Intestinal/microbiologia , Miofibroblastos/fisiologia , Técnicas de Cultura de Órgãos , Organoides/fisiologia , Prostaglandina D2/biossíntese , Transdução de Sinais , Receptor 4 Toll-Like/imunologia
15.
Nature ; 541(7636): 182-187, 2017 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-28052057

RESUMO

Despite the global prevalence of gastric disease, there are few adequate models in which to study the fundus epithelium of the human stomach. We differentiated human pluripotent stem cells (hPSCs) into gastric organoids containing fundic epithelium by first identifying and then recapitulating key events in embryonic fundus development. We found that disruption of Wnt/ß-catenin signalling in mouse embryos led to conversion of fundic to antral epithelium, and that ß-catenin activation in hPSC-derived foregut progenitors promoted the development of human fundic-type gastric organoids (hFGOs). We then used hFGOs to identify temporally distinct roles for multiple signalling pathways in epithelial morphogenesis and differentiation of fundic cell types, including chief cells and functional parietal cells. hFGOs are a powerful model for studying the development of the human fundus and the molecular bases of human gastric physiology and pathophysiology, and also represent a new platform for drug discovery.


Assuntos
Fundo Gástrico/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Animais , Padronização Corporal , Diferenciação Celular , Linhagem da Célula , Descoberta de Drogas/métodos , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Epitélio/embriologia , Epitélio/metabolismo , Feminino , Fundo Gástrico/citologia , Fundo Gástrico/embriologia , Proteínas de Homeodomínio/metabolismo , Humanos , Masculino , Camundongos , Organoides/citologia , Organoides/embriologia , Organoides/metabolismo , Células Parietais Gástricas/citologia , Células Parietais Gástricas/metabolismo , Células-Tronco Pluripotentes/citologia , Fatores de Transcrição SOXB1/metabolismo , Esferoides Celulares/citologia , Esferoides Celulares/metabolismo , Transativadores/metabolismo , Via de Sinalização Wnt/genética , beta Catenina/agonistas
16.
Dev Biol ; 420(2): 230-238, 2016 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-27287882

RESUMO

In vitro human pluripotent stem cell (hPSC) derived tissues are excellent models to study certain aspects of normal human development. Current research in the field of hPSC derived tissues reveals these models to be inherently fetal-like on both a morphological and gene expression level. In this review we briefly discuss current methods for differentiating lung and intestinal tissue from hPSCs into individual 3-dimensional units called organoids. We discuss how these methods mirror what is known about in vivo signaling pathways of the developing embryo. Additionally, we will review how the inherent immaturity of these models lends them to be particularly valuable in the study of immature human tissues in the clinical setting of premature birth. Human lung organoids (HLOs) and human intestinal organoids (HIOs) not only model normal development, but can also be utilized to study several important diseases of prematurity such as respiratory distress syndrome (RDS), bronchopulmonary dysplasia (BPD), and necrotizing enterocolitis (NEC).


Assuntos
Intestinos/embriologia , Pulmão/embriologia , Modelos Biológicos , Organoides/citologia , Organoides/embriologia , Células-Tronco Pluripotentes/citologia , Maturidade dos Órgãos Fetais , Feto/citologia , Feto/embriologia , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Intestinos/citologia , Pulmão/citologia , Organogênese , Medicina Regenerativa
17.
J Microsc ; 251(3): 212-23, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23432616

RESUMO

Mammary branching morphogenesis occurs over a period of weeks deep inside an adipocyte-rich stroma. The adipocytes contain light-scattering lipid droplets that limit the depth of penetration of visible light. Organotypic culture methods were developed to enable high-resolution optical monitoring of branching morphogenesis ex vivo. A challenge has been to identify the best culture conditions to model specific developmental events. We recently demonstrated that collagen I induces protrusive invasion in both normal and neoplastic mammary epithelium. In this study, we observed that the abundance of collagen I fibrils correlated strongly with invasive behaviour, even when the collagen I concentration was identical. We found that the extent of fibril assembly was experimentally manipulable by varying the incubation time at 4°C following pH neutralization. We next tested the capacity of collagen I fibrils to induce invasive behaviour when presented in combination with basement membrane proteins (Matrigel). We found that epithelial organoids in mixed gels of collagen I and basement membrane proteins exhibited more extensive branching morphogenesis but did not initiate protrusions into the matrix. Organoids in pure Matrigel produced many small epithelial buds that were bare of myoepithelial cells. Surprisingly, organoids in mixed gels of collagen I and Matrigel produced fewer epithelial buds, the buds elongated further, and the elongating buds remained covered by myoepithelial cells. Our mixed gels therefore provide a more physiologically accurate model of mammary branching morphogenesis. Our results also suggest that changes in the composition of the extracellular matrix could induce migration of epithelial cells past myoepithelial coverage.


Assuntos
Movimento Celular , Células Epiteliais/fisiologia , Matriz Extracelular/metabolismo , Glândulas Mamárias Animais/embriologia , Células Musculares/fisiologia , Animais , Colágeno Tipo I/metabolismo , Técnicas In Vitro , Camundongos , Imagem Óptica , Organoides/embriologia
18.
J Am Soc Nephrol ; 23(11): 1857-68, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23085631

RESUMO

The shortage of transplantable organs provides an impetus to develop tissue-engineered alternatives. Producing tissues similar to immature kidneys from simple suspensions of fully dissociated embryonic renal cells is possible in vitro, but glomeruli do not form in the avascular environment. Here, we constructed renal organoids from single-cell suspensions derived from E11.5 kidneys and then implanted these organoids below the kidney capsule of a living rat host. This implantation resulted in further maturation of kidney tissue, formation of vascularized glomeruli with fully differentiated capillary walls, including the slit diaphragm, and appearance of erythropoietin-producing cells. The implanted tissue exhibited physiologic functions, including tubular reabsorption of macromolecules, that gained access to the tubular lumen on glomerular filtration. The ability to generate vascularized nephrons from single-cell suspensions marks a significant step to the long-term goal of replacing renal function by a tissue-engineered kidney.


Assuntos
Rim/embriologia , Organoides/embriologia , Engenharia Tecidual/métodos , Animais , Órgãos Bioartificiais , Rim/irrigação sanguínea , Rim/citologia , Rim/fisiologia , Transplante de Rim/métodos , Masculino , Camundongos , Organoides/citologia , Organoides/transplante , Ratos , Ratos Nus , Transplante Heterólogo , Fator A de Crescimento do Endotélio Vascular/administração & dosagem
19.
Am J Physiol Cell Physiol ; 286(6): C1344-52, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-14960414

RESUMO

Using an embryoid body (EB) culture system, we have made a functional organlike cluster: the "gut" from embryonic stem (ES) cells (ES gut). There are many types of ES clusters, because ES cells have a pluripotent ability to develop into a wide range of cell types. Before inducing specific differentiation by exogenously added factors, we characterized comprehensive physiological and morphological properties of ES guts. Each ES gut has a hemispherical (or cystic) structure and exhibits spontaneous contractions [mean frequency: 13.5 +/- 8.8 cycles per min (cpm)]. A dense distribution of interstitial cells of Cajal (ICC) was identified by c-Kit immunoreactivity, and specific subcellular structures of ICC and smooth muscle cells were identified with electron microscopy. ICC frequently formed close contacts with the neighboring smooth muscle cells and occasionally formed gap junctions with other ICC. Widely propagating intracellular Ca(2+) concentration oscillations were generated in the ES gut from the aggregates of c-Kit immunopositive cells. Plateau potentials, possibly pacemaker potentials in ICC, and electrical slow waves were recorded for the first time. These events were nifedipine insensitive, as in the mouse gut. Our present results indicate that the rhythmic pacemaker activity generated in ICC efficiently spreads to smooth muscle cells and drives spontaneous rhythmic contractions of the ES gut. The present characterization of physiological and morphological properties of ES gut paves the way for making appropriate models to investigate the origin of rhythmicity in the gut.


Assuntos
Trato Gastrointestinal/embriologia , Organogênese/fisiologia , Organoides/embriologia , Células-Tronco Pluripotentes/metabolismo , Animais , Relógios Biológicos/efeitos dos fármacos , Relógios Biológicos/fisiologia , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Sinalização do Cálcio/fisiologia , Diferenciação Celular/fisiologia , Células Cultivadas , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/ultraestrutura , Junções Intercelulares/fisiologia , Junções Intercelulares/ultraestrutura , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Microscopia Eletrônica , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/ultraestrutura , Técnicas de Cultura de Órgãos/métodos , Organoides/metabolismo , Organoides/ultraestrutura , Peristaltismo/fisiologia , Células-Tronco Pluripotentes/ultraestrutura , Proteínas Proto-Oncogênicas c-kit/metabolismo
20.
Lab Invest ; 83(12): 1811-20, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14691299

RESUMO

Early embryonic blood vessels are typically composed of fragile tubes of endothelial cells encircled by vascular smooth muscle cells. Early human vasculogenesis was explored in spontaneous and directed differentiation models derived from human embryonic stem (HES) cells. In a 3-dimensional (3D) model, HES cells were studied for their potential for vascular differentiation during the spontaneous formation of embryoid bodies. Directed differentiation was investigated by means of a 2-dimensional (2D) differentiation method to promote vascular differentiation from HES cells (without the formation of embryoid bodies). Using this latter approach, up-regulation of early lineage markers of endothelial progenitors were induced. Additional culture under strict conditions and exposure to angiogenic growth factors resulted in a prolonged differentiation pathway into mature endothelial cells and up-regulation of vascular smooth muscle cell markers. The use of 3D collagen gels and Matrigel assays for the induction and inhibition of human vascular sprouting in vitro further established the vascular potential of the cells generated by the 2D differentiation system. Our study shows that HES cells can provide useful models to study early differentiation and development of blood vessels. Moreover, the 2D differentiation model facilitates both the production of vascular lineage cells from HES cells for various potential therapeutic applications and also provides a model for studying the mechanisms involved in early human embryonic blood vessel development.


Assuntos
Embrião de Mamíferos/citologia , Endotélio Vascular/embriologia , Músculo Liso Vascular/embriologia , Organogênese/fisiologia , Células-Tronco/citologia , Becaplermina , Diferenciação Celular , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Humanos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Organogênese/efeitos dos fármacos , Organoides/citologia , Organoides/efeitos dos fármacos , Organoides/embriologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Proto-Oncogênicas c-sis , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA