Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 745
Filtrar
1.
Nat Cell Biol ; 26(6): 868-877, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38849542

RESUMEN

Despite a distinct developmental origin, extraembryonic cells in mice contribute to gut endoderm and converge to transcriptionally resemble their embryonic counterparts. Notably, all extraembryonic progenitors share a non-canonical epigenome, raising several pertinent questions, including whether this landscape is reset to match the embryonic regulation and if extraembryonic cells persist into later development. Here we developed a two-colour lineage-tracing strategy to track and isolate extraembryonic cells over time. We find that extraembryonic gut cells display substantial memory of their developmental origin including retention of the original DNA methylation landscape and resulting transcriptional signatures. Furthermore, we show that extraembryonic gut cells undergo programmed cell death and neighbouring embryonic cells clear their remnants via non-professional phagocytosis. By midgestation, we no longer detect extraembryonic cells in the wild-type gut, whereas they persist and differentiate further in p53-mutant embryos. Our study provides key insights into the molecular and developmental fate of extraembryonic cells inside the embryo.


Asunto(s)
Apoptosis , Linaje de la Célula , Metilación de ADN , Endodermo , Regulación del Desarrollo de la Expresión Génica , Animales , Endodermo/citología , Endodermo/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Fagocitosis , Ratones Endogámicos C57BL , Ratones , Diferenciación Celular , Femenino , Desarrollo Embrionario , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Ratones Transgénicos , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/embriología , Tracto Gastrointestinal/metabolismo
2.
Cells ; 13(10)2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38786037

RESUMEN

Intestinal homeostasis results from the proper interplay among epithelial cells, the enteric nervous system (ENS), interstitial cells of Cajal (ICCs), smooth muscle cells, the immune system, and the microbiota. The disruption of this balance underpins the onset of gastrointestinal-related diseases. The scarcity of models replicating the intricate interplay between the ENS and the intestinal epithelium highlights the imperative for developing novel methods. We have pioneered a sophisticated tridimensional in vitro technique, coculturing small intestinal organoids with myenteric and submucosal neurons. Notably, we have made significant advances in (1) refining the isolation technique for culturing the myenteric plexus, (2) enhancing the isolation of the submucosal plexus-both yielding mixed cultures of enteric neurons and glial cells from both plexuses, and (3) subsequently co-culturing myenteric and submucosal neurons with small intestinal organoids. This co-culture system establishes neural innervations with intestinal organoids, allowing for the investigation of regulatory interactions in the context of gastrointestinal diseases. Furthermore, we have developed a method for microinjecting the luminal space of small intestinal organoids with fluorescently labeled compounds. This technique possesses broad applicability such as the assessment of intestinal permeability, transcytosis, and immunocytochemical and immunofluorescence applications. This microinjection method could be extended to alternative experimental setups, incorporating bacterial species, or applying treatments to study ENS-small intestinal epithelium interactions. Therefore, this technique serves as a valuable tool for evaluating the intricate interplay between neuronal and intestinal epithelial cells (IECs) and shows great potential for drug screening, gene editing, the development of novel therapies, the modeling of infectious diseases, and significant advances in regenerative medicine. The co-culture establishment process spans twelve days, making it a powerful asset for comprehensive research in this critical field.


Asunto(s)
Técnicas de Cocultivo , Intestino Delgado , Plexo Mientérico , Organoides , Animales , Ratones , Técnicas de Cocultivo/métodos , Tracto Gastrointestinal/inervación , Tracto Gastrointestinal/citología , Intestino Delgado/citología , Plexo Mientérico/citología , Neuronas/citología , Neuronas/metabolismo , Organoides/citología , Plexo Submucoso/citología
3.
Int J Biol Sci ; 20(7): 2476-2490, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38725863

RESUMEN

Peristaltic movements in gut are essential to propel ingested materials through the gastrointestinal tract. Intestinal resident macrophages play an important role in this physiological function through protecting enteric neurons. However, it is incompletely clear how individuals maintain the homeostasis of gut motility. Here we found that NLRP3 is a critical factor in controlling loss of muscularis resident macrophages (MMs), and demonstrate that MMs are involved in the homeostasis of excitatory neurons such as choline acetyltransferase (ChAT)+ and vesicular glutamate transporter 2 (VGLUT2)+ but not inhibitory neuronal nitric oxide synthase (nNOS)+ neurons. NLRP3 knockout (KO) mice had enhanced gut motility and increased neurons, especially excitatory ChAT+ and VGLUT2+ neurons. Single cell analyses showed that there had increased resident macrophages, especially MMs in NLRP3 KO mice. The MM proportion in the resident macrophages was markedly higher than those in wild-type (WT) or caspase 1/11 KO mice. Deletion of the MMs and transplantation of the NLRP3 KO bone marrow cells showed that survival of the gut excitatory ChAT+ and VGLUT2+ neurons was dependent on the MMs. Gut microbiota metabolites ß-hydroxybutyrate (BHB) could promote gut motility through protecting MMs from pyroptosis. Thus, our data suggest that MMs regulated by NLRP3 maintain the homeostasis of excitatory neurons.


Asunto(s)
Motilidad Gastrointestinal , Macrófagos , Proteína con Dominio Pirina 3 de la Familia NLR , Neuronas , Ratones , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Neuronas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Masculino , Femenino , Animales , Ratones Noqueados , Plexo Mientérico/citología , Plexo Mientérico/metabolismo , Colon/citología , Colon/metabolismo , Membrana Mucosa/citología , Membrana Mucosa/metabolismo
4.
Nature ; 607(7920): 762-768, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35794484

RESUMEN

Gastrointestinal health depends on the adaptive immune system tolerating the foreign proteins in food1,2. This tolerance is paradoxical because the immune system normally attacks foreign substances by generating inflammation. Here we addressed this conundrum by using a sensitive cell enrichment method to show that polyclonal CD4+ T cells responded to food peptides, including a natural one from gliadin, by proliferating weakly in secondary lymphoid organs of the gut-liver axis owing to the action of regulatory T cells. A few food-specific T cells then differentiated into T follicular helper cells that promoted a weak antibody response. Most cells in the expanded population, however, lacked canonical T helper lineage markers and fell into five subsets dominated by naive-like or T follicular helper-like anergic cells with limited capacity to form inflammatory T helper 1 cells. Eventually, many of the T helper lineage-negative cells became regulatory T cells themselves through an interleukin-2-dependent mechanism. Our results indicate that exposure to food antigens causes cognate CD4+ naive T cells to form a complex set of noncanonical hyporesponsive T helper cell subsets that lack the inflammatory functions needed to cause gut pathology and yet have the potential to produce regulatory T cells that may suppress it.


Asunto(s)
Linfocitos T CD4-Positivos , Alimentos , Tolerancia Inmunológica , Alérgenos/inmunología , Formación de Anticuerpos , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Proteínas en la Dieta/inmunología , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/inmunología , Gliadina/inmunología , Tolerancia Inmunológica/inmunología , Inflamación , Interleucina-2/inmunología , Hígado/citología , Hígado/inmunología , Tejido Linfoide/citología , Tejido Linfoide/inmunología , Fragmentos de Péptidos/inmunología , Células T Auxiliares Foliculares/citología , Células T Auxiliares Foliculares/inmunología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Células TH1/citología , Células TH1/inmunología
5.
Int. j. morphol ; 40(1): 242-250, feb. 2022. ilus, tab
Artículo en Inglés | LILACS | ID: biblio-1385577

RESUMEN

SUMMARY: In the present study, we aimed to determine the localization and distribution of entero-endocrine cells in the gastrointestinal tract by immunohistochemical methods and understand the structure of the glycoproteins elaborated by the epithelium the digestive tract regions by histochemical methods. The nine sparrowhawks were euthanized, and gastrointestinal tract tissues were removed and fixed in formalin. The gastrointestinal tract sections were stained with immunohistochemical and histochemical techniques to evaluate the enteroendocrine cells and histomorphometric analysis. The results showed that the numbers of somatostatin in the ventriculus, gastrin in the proventriculus, serotonin in the duodenum and jejunum immunopositivity are higher, remaining segments of the gastrointestinal tract are detected slight positivity in the glucagon, gastrin, serotonin, and somatostatin. In conclusion, some endocrine cells localization and distribution and histomorphometry, and goblet cell counts were revealed in the gastrointestinal tract of the sparrowhawks.


RESUMEN: El objetivo del presente estudio fue determinar la localización y distribución de células enteroendocrinas en el tracto gastrointestinal de gavilán, a través de métodos inmunohistoquímicos y comprender la estructura de las glicoproteínas elaboradas por el epitelio de las regiones del tracto digestivo. Se sacrificaron nueve gavilanes y los tejidos del tracto gastrointestinal se extrajeron y se fijaron en formalina. Las secciones del tracto gastrointestinal se tiñeron con técnicas inmunohistoquímicas e histoquímicas para evaluar las células enteroendocrinas y se realizó análisis histo-morfométrico. Los re- sultados indicaron que los números de inmunopositividad de somatostatina en el ventrículo, gastrina en el proventrículo, serotonina en el duodeno y yeyuno son más altos, en los segmentos restantes del tracto gastrointestinal, se detecta además una ligera positividad de glucagón, gastrina, serotonina y somatostatina. En conclusión en el tracto gastrointestinal de gavilán se observó cierta localización y distribución de células endocrinas e histomorfometría, y recuentos de células caliciformes.


Asunto(s)
Animales , Células Enteroendocrinas , Tracto Gastrointestinal/citología , Falconiformes , Inmunohistoquímica
6.
Cells ; 11(2)2022 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-35053422

RESUMEN

A number of gut epithelial cells derived immunological factors such as cytokines and chemokines, which are stimulated by the gut microbiota, can regulate host immune responses to maintain a well-balance between gut microbes and host immune system. Multiple specialized immune cell populations, such as macrophages, dendritic cells (DCs), innate lymphoid cells, and T regulatory (Treg) cells, can communicate with intestinal epithelial cells (IEC) and/or the gut microbiota bi-directionally. The gut microbiota contributes to the differentiation and function of resident macrophages. Situated at the interface between the gut commensals and macrophages, the gut epithelium is crucial for gut homeostasis in microbial recognition, signaling transformation, and immune interactions, apart from being a physical barrier. Thus, three distinct but interactive components-macrophages, microbiota, and IEC-can form a network for the delicate and dynamic regulation of intestinal homeostasis. In this review, we will discuss the crucial features of gut microbiota, macrophages, and IEC. We will also summarize recent advances in understanding the cooperative and dynamic interactions among the gut microbiota, gut macrophages, and IEC, which constitute a special network for gut homeostasis.


Asunto(s)
Células Epiteliales/citología , Microbioma Gastrointestinal , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/microbiología , Homeostasis , Macrófagos/citología , Animales , Humanos , Modelos Biológicos
7.
Cells ; 10(11)2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34831120

RESUMEN

Medullary thyroid carcinoma contributes to about 3-4% of thyroid cancers and affects C cells rather than follicular cells. Thyroid C cell differentiation from human pluripotent stem cells has not been reported. We report the stepwise differentiation of human embryonic stem cells into thyroid C cell-like cells through definitive endoderm and anterior foregut endoderm and ultimobranchial body-like intermediates in monolayer and 3D Matrigel culture conditions. The protocol involved sequential treatment with interferon/transferrin/selenium/pyruvate, foetal bovine serum, and activin A, then IGF-1 (Insulin-like growth factor 1), on the basis of embryonic thyroid developmental sequence. As well as expressing C cell lineage relative to follicular-lineage markers by qPCR (quantitative polymerase chain reaction) and immunolabelling, these cells by ELISA (enzyme-linked immunoassay) exhibited functional properties in vitro of calcitonin storage and release of calcitonin on calcium challenge. This method will contribute to developmental studies of the human thyroid gland and facilitate in vitro modelling of medullary thyroid carcinoma and provide a valuable platform for drug screening.


Asunto(s)
Células Madre Pluripotentes/citología , Glándula Tiroides/citología , Andamios del Tejido/química , Biomarcadores/metabolismo , Calcitonina/metabolismo , Calcio/metabolismo , Diferenciación Celular/efectos de los fármacos , Colágeno/farmacología , Combinación de Medicamentos , Endodermo/citología , Tracto Gastrointestinal/citología , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/efectos de los fármacos , Células Madre Embrionarias Humanas/metabolismo , Humanos , Laminina/farmacología , Sistemas Neurosecretores/citología , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Proteoglicanos/farmacología
8.
Front Immunol ; 12: 745332, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34671359

RESUMEN

The induction of trained immunity represents an emerging concept defined as the ability of innate immune cells to acquire a memory phenotype, which is a typical hallmark of the adaptive response. Key points modulated during the establishment of trained immunity include epigenetic, metabolic and functional changes in different innate-immune and non-immune cells. Regarding to epigenetic changes, it has been described that long non-coding RNAs (LncRNAs) act as molecular scaffolds to allow the assembly of chromatin-remodeling complexes that catalyze epigenetic changes on chromatin. On the other hand, relevant metabolic changes that occur during this process include increased glycolytic rate and the accumulation of metabolites from the tricarboxylic acid (TCA) cycle, which subsequently regulate the activity of histone-modifying enzymes that ultimately drive epigenetic changes. Functional consequences of established trained immunity include enhanced cytokine production, increased antigen presentation and augmented antimicrobial responses. In this article, we will discuss the current knowledge regarding the ability of different cell subsets to acquire a trained immune phenotype and the molecular mechanisms involved in triggering such a response. This knowledge will be helpful for the development of broad-spectrum therapies against infectious diseases based on the modulation of epigenetic and metabolic cues regulating the development of trained immunity.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Inmunidad Celular , Inmunidad Innata/inmunología , Memoria Inmunológica/inmunología , Inmunidad Adaptativa/genética , Inmunidad Adaptativa/inmunología , Inmunidad Adaptativa/fisiología , Animales , Vacuna BCG/inmunología , Bronquios/citología , Bronquios/inmunología , Citocinas/fisiología , Metabolismo Energético , Epigénesis Genética , Células Epiteliales/inmunología , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/inmunología , Células Madre Hematopoyéticas/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/fisiología , Humanos , Inmunidad Celular/genética , Inmunidad Celular/fisiología , Inmunidad Innata/genética , Inmunidad Innata/fisiología , Memoria Inmunológica/genética , Memoria Inmunológica/fisiología , Linfocitos/inmunología , Ratones , Células Mieloides/inmunología , NAD/fisiología , Piel/citología , Piel/inmunología
9.
Elife ; 102021 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-34553686

RESUMEN

The gut is the primary interface between an animal and food, but how it adapts to qualitative dietary variation is poorly defined. We find that the Drosophila midgut plastically resizes following changes in dietary composition. A panel of nutrients collectively promote gut growth, which sugar opposes. Diet influences absolute and relative levels of enterocyte loss and stem cell proliferation, which together determine cell numbers. Diet also influences enterocyte size. A high sugar diet inhibits translation and uncouples intestinal stem cell proliferation from expression of niche-derived signals, but, surprisingly, rescuing these effects genetically was not sufficient to modify diet's impact on midgut size. However, when stem cell proliferation was deficient, diet's impact on enterocyte size was enhanced, and reducing enterocyte-autonomous TOR signaling was sufficient to attenuate diet-dependent midgut resizing. These data clarify the complex relationships between nutrition, epithelial dynamics, and cell size, and reveal a new mode of plastic, diet-dependent organ resizing.


Asunto(s)
Dieta , Drosophila melanogaster/crecimiento & desarrollo , Tracto Gastrointestinal/crecimiento & desarrollo , Animales , Animales Modificados Genéticamente , Proliferación Celular , Drosophila melanogaster/fisiología , Enterocitos/citología , Femenino , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/fisiología , Masculino , Nicho de Células Madre
10.
J Insect Physiol ; 134: 104309, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34496279

RESUMEN

The adult Drosophila intestinal epithelium must be tightly regulated to maintain regeneration and homeostasis. The dysregulation of the regenerative capacity is frequently associated with intestinal diseases such as inflammation and tumorigenesis. Here, we showed that the G protein-coupled receptor Anchor maintains Drosophila adult midgut homeostasis by restricting Jun-N-terminal kinase (JNK) and Notch pathway activity. anchor inactivation resulted in aberrant JNK pathway activation, which led to excessive enteroblast (EB) production and premature enterocyte (EC) differentiation. In addition, increased Notch levels promoted premature EC differentiation following the loss of anchor. This defect induced by the loss of anchor ultimately caused sensitivity to stress or environmental challenge in adult flies. Taken together, our results demonstrate that the activity of anchor is essential to coordinate stem cell differentiation and proliferation to maintain intestinal homeostasis.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Homeostasis/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Animales , Proliferación Celular , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Drosophila melanogaster/fisiología , Enterocitos , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/metabolismo , Sistema de Señalización de MAP Quinasas , Receptores Notch/metabolismo , Transducción de Señal
11.
J Med Virol ; 93(12): 6671-6685, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34324210

RESUMEN

Infection by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes a wide spectrum of syndromes involving multiple organ systems and is primarily mediated by viral spike (S) glycoprotein through the receptor-binding domain (RBD) and numerous cellular proteins including ACE2, transmembrane serine protease 2 (TMPRSS2), kidney injury molecule-1 (Kim-1), and neuropilin-1 (NRP-1). In this study, we examined the entry tropism of SARS-CoV-2 and SARS-CoV using S protein-based pseudoviruses to infect 22 cell lines and 3 types of primary cells isolated from respiratory, urinary, digestive, reproductive, and immune systems. At least one cell line or type of primary cell from each organ system was infected by both pseudoviruses. Infection by pseudoviruses is effectively blocked by S1, RBD, and ACE2 recombinant proteins, and more weakly by Kim-1 and NRP-1 recombinant proteins. Furthermore, cells with robust SARS-CoV-2 pseudovirus infection had strong expression of either ACE2 or Kim-1 and NRP-1 proteins. ACE2 glycosylation appeared to be critical for the infections of both viruses as there was a positive correlation between infectivity of either SARS-CoV-2 or SARS-CoV pseudovirus with the level of glycosylated ACE2 (gly-ACE2). These results reveal that SARS-CoV-2 cell entry could be mediated by either an ACE2-dependent or -independent mechanism, thus providing a likely molecular basis for its broad tropism for a wide variety of cell types.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Tracto Gastrointestinal/virología , Genitales/virología , Receptor Celular 1 del Virus de la Hepatitis A/metabolismo , Sistema Inmunológico/virología , Neuropilina-1/metabolismo , Sistema Respiratorio/virología , SARS-CoV-2/fisiología , Serina Endopeptidasas/metabolismo , Internalización del Virus , Western Blotting , COVID-19/metabolismo , COVID-19/virología , Línea Celular , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Tracto Gastrointestinal/citología , Genitales/citología , Humanos , Sistema Inmunológico/citología , Sistema Respiratorio/citología
12.
Microbiol Spectr ; 9(1): e0000321, 2021 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-34106568

RESUMEN

Gastrointestinal infections cause significant morbidity and mortality worldwide. The complexity of human biology and limited insights into host-specific infection mechanisms are key barriers to current therapeutic development. Here, we demonstrate that two-dimensional epithelial monolayers derived from human intestinal organoids, combined with in vivo-like bacterial culturing conditions, provide significant advancements for the study of enteropathogens. Monolayers from the terminal ileum, cecum, and ascending colon recapitulated the composition of the gastrointestinal epithelium, in which several techniques were used to detect the presence of enterocytes, mucus-producing goblet cells, and other cell types following differentiation. Importantly, the addition of receptor activator of nuclear factor kappa-B ligand (RANKL) increased the presence of M cells, critical antigen-sampling cells often exploited by enteric pathogens. For infections, bacteria were grown under in vivo-like conditions known to induce virulence. Overall, interesting patterns of tissue tropism and clinical manifestations were observed. Shigella flexneri adhered efficiently to the cecum and colon; however, invasion in the colon was best following RANKL treatment. Both Salmonella enterica serovars Typhi and Typhimurium displayed different infection patterns, with S. Typhimurium causing more destruction of the terminal ileum and S. Typhi infecting the cecum more efficiently than the ileum, particularly with regard to adherence. Finally, various pathovars of Escherichia coli validated the model by confirming only adherence was observed with these strains. This work demonstrates that the combination of human-derived tissue with targeted bacterial growth conditions enables powerful analyses of human-specific infections that could lead to important insights into pathogenesis and accelerate future vaccine development. IMPORTANCE While traditional laboratory techniques and animal models have provided valuable knowledge in discerning virulence mechanisms of enteric pathogens, the complexity of the human gastrointestinal tract has hindered our understanding of physiologically relevant, human-specific interactions; and thus, has significantly delayed successful vaccine development. The human intestinal organoid-derived epithelial monolayer (HIODEM) model closely recapitulates the diverse cell populations of the intestine, allowing for the study of human-specific infections. Differentiation conditions permit the expansion of various cell populations, including M cells that are vital to immune recognition and the establishment of infection by some bacteria. We provide details of reproducible culture methods and infection conditions for the analyses of Shigella, Salmonella, and pathogenic Escherichia coli in which tissue tropism and pathogen-specific infection patterns were detected. This system will be vital for future studies that explore infection conditions, health status, or epigenetic differences and will serve as a novel screening platform for therapeutic development.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Infecciones por Enterobacteriaceae/microbiología , Enterobacteriaceae/fisiología , Tracto Gastrointestinal/microbiología , Organoides/microbiología , Enterobacteriaceae/genética , Enterobacteriaceae/patogenicidad , Enterocitos/microbiología , Células Epiteliales/citología , Células Epiteliales/microbiología , Epitelio/microbiología , Tracto Gastrointestinal/citología , Humanos , Organoides/citología , Virulencia
13.
Front Immunol ; 12: 646633, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33679811

RESUMEN

Conjugation with the small ubiquitin-like modifier (SUMO) constitutes a key post-translational modification regulating the stability, activity, and subcellular localization of its target proteins. However, the vast numbers of identified SUMO substrates obscure a clear view on the function of SUMOylation in health and disease. This article presents a comprehensive review on the physiological relevance of SUMOylation by discussing how global SUMOylation levels-rather than specific protein SUMOylation-shapes the immune response. In particular, we highlight the growing body of work on SUMOylation in intestinal pathologies, because of the unique metabolic, infectious, and inflammatory challenges of this organ. Recent studies show that global SUMOylation can help restrain detrimental inflammation while maintaining immune defenses and tissue integrity. These results warrant further efforts to develop new therapeutic tools and strategies to control SUMOylation in infectious and inflammatory disorders.


Asunto(s)
Tracto Gastrointestinal/inmunología , Inflamación/inmunología , Procesamiento Proteico-Postraduccional/inmunología , Estrés Fisiológico/inmunología , Animales , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/metabolismo , Humanos , Interferones/inmunología , Interferones/metabolismo , Macrófagos/inmunología , Neutrófilos/inmunología , Sumoilación/inmunología
14.
Int J Mol Sci ; 22(5)2021 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-33652988

RESUMEN

In this Review, we briefly describe the basic virology and pathogenesis of SARS-CoV-2, highlighting how stem cell technology and organoids can contribute to the understanding of SARS-CoV-2 cell tropisms and the mechanism of disease in the human host, supporting and clarifying findings from clinical studies in infected individuals. We summarize here the results of studies, which used these technologies to investigate SARS-CoV-2 pathogenesis in different organs. Studies with in vitro models of lung epithelia showed that alveolar epithelial type II cells, but not differentiated lung alveolar epithelial type I cells, are key targets of SARS-CoV-2, which triggers cell apoptosis and inflammation, while impairing surfactant production. Experiments with human small intestinal organoids and colonic organoids showed that the gastrointestinal tract is another relevant target for SARS-CoV-2. The virus can infect and replicate in enterocytes and cholangiocytes, inducing cell damage and inflammation. Direct viral damage was also demonstrated in in vitro models of human cardiomyocytes and choroid plexus epithelial cells. At variance, endothelial cells and neurons are poorly susceptible to viral infection, thus supporting the hypothesis that neurological symptoms and vascular damage result from the indirect effects of systemic inflammatory and immunological hyper-responses to SARS-CoV-2 infection.


Asunto(s)
COVID-19/patología , Organoides/virología , SARS-CoV-2/fisiología , Células Madre/virología , Animales , Apoptosis , COVID-19/virología , Sistema Cardiovascular/citología , Sistema Cardiovascular/patología , Sistema Cardiovascular/virología , Sistema Nervioso Central/citología , Sistema Nervioso Central/patología , Sistema Nervioso Central/virología , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/patología , Tracto Gastrointestinal/virología , Humanos , Inflamación/patología , Inflamación/virología , Pulmón/citología , Pulmón/patología , Pulmón/virología , Organoides/patología , Células Madre/patología , Tropismo Viral , Internalización del Virus
15.
J Mol Med (Berl) ; 99(4): 517-530, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33538854

RESUMEN

The human gastrointestinal tract is in constant contact with microbial stimuli. Its barriers have to ensure co-existence with the commensal bacteria, while enabling surveillance of intruding pathogens. At the centre of the interaction lies the epithelial layer, which marks the boundaries of the body. It is equipped with a multitude of different innate immune sensors, such as Toll-like receptors, to mount inflammatory responses to microbes. Dysfunction of this intricate system results in inflammation-associated pathologies, such as inflammatory bowel disease. However, the complexity of the cellular interactions, their molecular basis and their development remains poorly understood. In recent years, stem cell-derived organoids have gained increasing attention as promising models for both development and a broad range of pathologies, including infectious diseases. In addition, organoids enable the study of epithelial innate immunity in vitro. In this review, we focus on the gastrointestinal epithelial barrier and its regional organization to discuss innate immune sensing and development.


Asunto(s)
Células Epiteliales/inmunología , Tracto Gastrointestinal/inmunología , Inmunidad Innata , Organoides , Adulto , Animales , Bancos de Muestras Biológicas , Polaridad Celular , Predicción , Microbioma Gastrointestinal/inmunología , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología , Perfilación de la Expresión Génica , Células Caliciformes/inmunología , Humanos , Tolerancia Inmunológica , Recién Nacido , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/inmunología , Ratones , Modelos Inmunológicos , FN-kappa B/fisiología , Especificidad de Órganos , Organoides/citología , Organoides/inmunología , Células de Paneth/inmunología , Ganglios Linfáticos Agregados/inmunología , Células Madre/inmunología , Receptores Toll-Like/inmunología
16.
Cancer Lett ; 504: 116-124, 2021 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-33577978

RESUMEN

Tissue engineered organoids are simple biomodels that can emulate the structural and functional complexity of specific organs. Here, we review developments in three-dimensional (3D) artificial cell constructs to model gastrointestinal dynamics towards cancer diagnosis. We describe bottom-up approaches to fabricate close-packed cell aggregates, from the use of biochemical and physical cues to guide the self-assembly of organoids, to the use of engineering approaches, including 3D printing/additive manufacturing and external field-driven protocols. Finally, we outline the main challenges and possible risks regarding the potential translation of gastrointestinal organoids from laboratory settings to patient-specific models in clinical applications.


Asunto(s)
Tracto Gastrointestinal/citología , Modelos Biológicos , Organoides/citología , Medicina de Precisión , Ingeniería de Tejidos/métodos , Diferenciación Celular , Humanos
17.
J Mol Med (Berl) ; 99(4): 555-568, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33459801

RESUMEN

Three-dimensional organoids have been widely used for developmental and disease modeling. Organoids are derived from both adult and pluripotent stem cells. Various types are available for mimicking almost all major organs and tissues in the mouse and human. While culture protocols for stepwise differentiation and long-term expansion are well established, methods for genetic manipulation in organoids still need further standardization. In this review, we summarized different methods for organoid genetics and provide the pros and cons of each method for designing an optimal strategy.


Asunto(s)
Ingeniería Genética/métodos , Organoides , Células Madre Adultas/citología , Células Madre Adultas/efectos de los fármacos , Animales , Animales Modificados Genéticamente , Encéfalo/citología , Sistemas CRISPR-Cas , Técnicas de Reprogramación Celular , Tracto Gastrointestinal/citología , Edición Génica/métodos , Técnicas de Transferencia de Gen , Ingeniería Genética/tendencias , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Riñón/citología , Hígado/citología , Glándulas Mamarias Animales/citología , Ratones , Organoides/citología
18.
Expert Rev Gastroenterol Hepatol ; 15(1): 41-50, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32955375

RESUMEN

INTRODUCTION: The new Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is the etiologic agent of coronavirus disease 2019. Some authors reported pieces of evidence that patients with SARS-CoV-2 infection could have direct involvement of the gastrointestinal tract, and in symptomatic cases, gastrointestinal symptoms (diarrhea, nausea/vomiting, abdominal pain) could be very common. AREA COVERED: In this article, we reviewed current-published data of the gastrointestinal aspects involved in SARS-CoV-2 infection, including prevalence and incidence of specific symptoms, the presumptive biological mechanism of GI infection, prognosis, clinical management, and public health-related concerns on the possible risk of oral-fecal transmission. EXPERT OPINION: Different clues point to direct virus infection and replication in mucosal cells of the gastrointestinal tract. In vitro studies showed that SARS-CoV-2 could enter into the gastrointestinal epithelial cells by the Angiotensin-Converting enzyme two membrane receptor. These findings, coupled with the identification of viral RNA found in stools of patients, clearly suggest that direct involvement of the gastrointestinal tract is very likely. This can justify most of the gastrointestinal symptoms but also suggest a risk for an oral-fecal route for transmission, additionally or alternatively to the main respiratory route.


Asunto(s)
COVID-19/complicaciones , Enfermedades Gastrointestinales/epidemiología , Enfermedades Gastrointestinales/virología , ARN Viral/análisis , SARS-CoV-2/fisiología , Dolor Abdominal/epidemiología , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/transmisión , Diarrea/epidemiología , Células Epiteliales/metabolismo , Heces/química , Tracto Gastrointestinal/citología , Humanos , Incidencia , Náusea/epidemiología , Prevalencia , SARS-CoV-2/aislamiento & purificación , Glicoproteína de la Espiga del Coronavirus/metabolismo , Acoplamiento Viral , Vómitos/epidemiología
19.
Nat Mater ; 20(1): 22-29, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32958879

RESUMEN

Bioprinting promises enormous control over the spatial deposition of cells in three dimensions1-7, but current approaches have had limited success at reproducing the intricate micro-architecture, cell-type diversity and function of native tissues formed through cellular self-organization. We introduce a three-dimensional bioprinting concept that uses organoid-forming stem cells as building blocks that can be deposited directly into extracellular matrices conducive to spontaneous self-organization. By controlling the geometry and cellular density, we generated centimetre-scale tissues that comprise self-organized features such as lumens, branched vasculature and tubular intestinal epithelia with in vivo-like crypts and villus domains. Supporting cells were deposited to modulate morphogenesis in space and time, and different epithelial cells were printed sequentially to mimic the organ boundaries present in the gastrointestinal tract. We thus show how biofabrication and organoid technology can be merged to control tissue self-organization from millimetre to centimetre scales, opening new avenues for drug discovery, diagnostics and regenerative medicine.


Asunto(s)
Bioimpresión/métodos , Organoides/metabolismo , Matriz Extracelular/metabolismo , Tracto Gastrointestinal/citología
20.
J Dairy Sci ; 104(1): 1175-1182, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33162086

RESUMEN

The objective of this study was to compare the transcription of gene markers for gastrointestinal (GI) epithelial cells, including fatty acid binding protein 2 (FABP2) and cytokeratin 8 (KRT8), and tight junction complex genes (TJP1, CLDN1, CLDN4) in fecal RNA against several GI tract tissue sections in dairy calves. Eight healthy Jersey calves were euthanized at 5 wk of age, and postmortem samples were collected from rumen, duodenum, jejunum, ileum, large intestine, cecum, and feces for total RNA isolation. Tissues and fecal samples were immediately frozen in liquid nitrogen until RNA isolation. A real-time quantitative PCR analysis was performed using a single standard curve composited of equal amounts of all samples, including cDNA from fecal and GI tract tissues. The mRNA expression of the tight junctions TJP1, CLDN1, and CLDN4 was greater in fecal RNA compared with lower GI tract tissues (i.e., duodenum, jejunum, ileum, large intestine, and cecum). Similar to fecal RNA, rumen tissue had greater expression of tight junctions CLDN1 and CLDN4 than lower GI tract tissues. Similarly, rumen tissue had greater expression of TPJ1 than all lower GI tract tissues except duodenum. The expression of TJP1 and CLDN4 was greater in fecal RNA than in rumen tissue; in contrast, CLDN1 mRNA expression was greater in rumen tissue than in the fecal RNA. The expression of FABP2 was greater in duodenum in comparison to all tissue except ileum. The mRNA expression of FABP2 in fecal samples was similar to jejunum and ileum. The expression of KRT8 in fecal samples was similar to duodenum, large intestine, and cecum. The fecal RNA had a greater expression of KRT8 in comparison to jejunum and ileum. The rumen tissue had the lowest mRNA expression of KRT8. The expression levels of FABP2, KRT8, and tight junction genes observed in fecal transcripts suggest that a considerable amount of RNA derived from GI tract epithelial cells can be detected in fecal RNA, which is in agreement with previous data in neonatal dairy calves and other biological models including humans, rodents, and primates. The greater expression of tight junctions in fecal RNA in comparison to sections of the low GI remains to be understood, and due to the importance of tight junctions in GI physiology, further clarification of this effect is warranted. The similarities in mRNA expression of FABP2 and KRT8 between fecal RNA and intestinal sections add up to the accumulating evidence that fecal RNA can be used to investigate molecular alterations in the GI tract of neonatal dairy calves. Further research in this area should include high-throughput transcriptomic analysis via RNA-seq to uncover novel molecular markers for specific sections of the GI tract of neonates.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Biomarcadores/metabolismo , Bovinos/metabolismo , Tracto Gastrointestinal/metabolismo , Mucosa Intestinal/metabolismo , ARN/metabolismo , Animales , Bovinos/anatomía & histología , Ciego/metabolismo , Células Epiteliales/metabolismo , Heces , Tracto Gastrointestinal/citología , Íleon/metabolismo , Mucosa Intestinal/citología , Intestino Grueso , Yeyuno/metabolismo , Masculino , Reacción en Cadena en Tiempo Real de la Polimerasa , Rumen/metabolismo , Uniones Estrechas , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...