Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 752
Filtrar
Mais filtros

Eixos temáticos
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 180(4): 764-779.e20, 2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-32059779

RESUMO

The heterogeneity of endothelial cells (ECs) across tissues remains incompletely inventoried. We constructed an atlas of >32,000 single-EC transcriptomes from 11 mouse tissues and identified 78 EC subclusters, including Aqp7+ intestinal capillaries and angiogenic ECs in healthy tissues. ECs from brain/testis, liver/spleen, small intestine/colon, and skeletal muscle/heart pairwise expressed partially overlapping marker genes. Arterial, venous, and lymphatic ECs shared more markers in more tissues than did heterogeneous capillary ECs. ECs from different vascular beds (arteries, capillaries, veins, lymphatics) exhibited transcriptome similarity across tissues, but the tissue (rather than the vessel) type contributed to the EC heterogeneity. Metabolic transcriptome analysis revealed a similar tissue-grouping phenomenon of ECs and heterogeneous metabolic gene signatures in ECs between tissues and between vascular beds within a single tissue in a tissue-type-dependent pattern. The EC atlas taxonomy enabled identification of EC subclusters in public scRNA-seq datasets and provides a powerful discovery tool and resource value.


Assuntos
Células Endoteliais/metabolismo , Análise de Célula Única , Transcriptoma , Animais , Encéfalo/citologia , Sistema Cardiovascular/citologia , Células Endoteliais/classificação , Células Endoteliais/citologia , Trato Gastrointestinal/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculos/citologia , Especificidade de Órgãos , RNA-Seq , Testículo/citologia
2.
Cell ; 169(4): 570-586, 2017 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-28475890

RESUMO

Choices have consequences. Immune cells survey and migrate throughout the body and sometimes take residence in niche environments with distinct communities of cells, extracellular matrix, and nutrients that may differ from those in which they matured. Imbedded in immune cell physiology are metabolic pathways and metabolites that not only provide energy and substrates for growth and survival, but also instruct effector functions, differentiation, and gene expression. This review of immunometabolism will reference the most recent literature to cover the choices that environments impose on the metabolism and function of immune cells and highlight their consequences during homeostasis and disease.


Assuntos
Leucócitos/citologia , Leucócitos/imunologia , Animais , Trato Gastrointestinal/citologia , Trato Gastrointestinal/imunologia , Humanos , Leucócitos/metabolismo , Linfócitos T/imunologia , Microambiente Tumoral
3.
Cell ; 168(5): 928-943.e11, 2017 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-28215708

RESUMO

Within the human gut reside diverse microbes coexisting with the host in a mutually advantageous relationship. Evidence has revealed the pivotal role of the gut microbiota in shaping the immune system. To date, only a few of these microbes have been shown to modulate specific immune parameters. Herein, we broadly identify the immunomodulatory effects of phylogenetically diverse human gut microbes. We monocolonized mice with each of 53 individual bacterial species and systematically analyzed host immunologic adaptation to colonization. Most microbes exerted several specialized, complementary, and redundant transcriptional and immunomodulatory effects. Surprisingly, these were independent of microbial phylogeny. Microbial diversity in the gut ensures robustness of the microbiota's ability to generate a consistent immunomodulatory impact, serving as a highly important epigenetic system. This study provides a foundation for investigation of gut microbiota-host mutualism, highlighting key players that could identify important therapeutics.


Assuntos
Bactérias/classificação , Microbioma Gastrointestinal , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Imunidade Adaptativa , Animais , Fenômenos Fisiológicos Bacterianos , Trato Gastrointestinal/citologia , Trato Gastrointestinal/fisiologia , Vida Livre de Germes , Humanos , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Simbiose
4.
Cell ; 170(1): 185-198.e16, 2017 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-28648659

RESUMO

Dietary, microbial, and inflammatory factors modulate the gut-brain axis and influence physiological processes ranging from metabolism to cognition. The gut epithelium is a principal site for detecting such agents, but precisely how it communicates with neural elements is poorly understood. Serotonergic enterochromaffin (EC) cells are proposed to fulfill this role by acting as chemosensors, but understanding how these rare and unique cell types transduce chemosensory information to the nervous system has been hampered by their paucity and inaccessibility to single-cell measurements. Here, we circumvent this limitation by exploiting cultured intestinal organoids together with single-cell measurements to elucidate intrinsic biophysical, pharmacological, and genetic properties of EC cells. We show that EC cells express specific chemosensory receptors, are electrically excitable, and modulate serotonin-sensitive primary afferent nerve fibers via synaptic connections, enabling them to detect and transduce environmental, metabolic, and homeostatic information from the gut directly to the nervous system.


Assuntos
Células Quimiorreceptoras/metabolismo , Células Enterocromafins/metabolismo , Trato Gastrointestinal/citologia , Vias Neurais , Sequência de Aminoácidos , Animais , Sequência de Bases , Canais de Cálcio/metabolismo , Catecolaminas/metabolismo , Perfilação da Expressão Gênica , Humanos , Síndrome do Intestino Irritável/patologia , Camundongos , Fibras Nervosas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores Odorantes/metabolismo , Receptores 5-HT3 de Serotonina/metabolismo , Serotonina/metabolismo , Transdução de Sinais , Sinapses/metabolismo , Canal de Cátion TRPA1 , Canais de Potencial de Receptor Transitório/metabolismo
5.
Cell ; 161(1): 176-176.e1, 2015 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-25815994

RESUMO

The endoderm germ layer contributes to the respiratory and gastrointestinal (GI) lineages during development, giving rise to an array of specialized epithelial cell types lining organs, including the thyroid, thymus, lungs, liver, biliary system, pancreas, and intestines. This SnapShot timelines and summarizes key stages following gastrulation, including endoderm patterning, organ specification, and organogenesis. A lineage tree of the developing endocrine pancreas is outlined to further illustrate this process.


Assuntos
Trato Gastrointestinal/embriologia , Animais , Trato Gastrointestinal/citologia , Trato Gastrointestinal/metabolismo , Humanos , Organogênese , Pâncreas/citologia , Pâncreas/embriologia , Pâncreas/metabolismo , Fatores de Transcrição/metabolismo
6.
Cell ; 158(2): 239-240, 2014 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-25036623

RESUMO

Gastrointestinal motility causes movement of food during digestion through contractions of the gut smooth muscle. The enteric nervous system regulates these events, and Muller et al. now find that its interaction with the immune system, in concert with gut microbiota, provides an additional layer of regulation to this complex task.


Assuntos
Motilidade Gastrointestinal , Trato Gastrointestinal/citologia , Trato Gastrointestinal/microbiologia , Macrófagos/metabolismo , Animais
7.
Cell ; 158(2): 300-313, 2014 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-25036630

RESUMO

Intestinal peristalsis is a dynamic physiologic process influenced by dietary and microbial changes. It is tightly regulated by complex cellular interactions; however, our understanding of these controls is incomplete. A distinct population of macrophages is distributed in the intestinal muscularis externa. We demonstrate that, in the steady state, muscularis macrophages regulate peristaltic activity of the colon. They change the pattern of smooth muscle contractions by secreting bone morphogenetic protein 2 (BMP2), which activates BMP receptor (BMPR) expressed by enteric neurons. Enteric neurons, in turn, secrete colony stimulatory factor 1 (CSF1), a growth factor required for macrophage development. Finally, stimuli from microbial commensals regulate BMP2 expression by macrophages and CSF1 expression by enteric neurons. Our findings identify a plastic, microbiota-driven crosstalk between muscularis macrophages and enteric neurons that controls gastrointestinal motility. PAPERFLICK:


Assuntos
Motilidade Gastrointestinal , Trato Gastrointestinal/citologia , Trato Gastrointestinal/microbiologia , Macrófagos/metabolismo , Animais , Proteína Morfogenética Óssea 2/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Trato Gastrointestinal/inervação , Trato Gastrointestinal/fisiologia , Técnicas In Vitro , Fator Estimulador de Colônias de Macrófagos , Camundongos , Neurônios/metabolismo , Peristaltismo , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Transdução de Sinais
8.
Nature ; 607(7920): 762-768, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35794484

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos , Alimentos , Tolerância Imunológica , Alérgenos/imunologia , Formação de Anticorpos , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Proteínas Alimentares/imunologia , Trato Gastrointestinal/citologia , Trato Gastrointestinal/imunologia , Gliadina/imunologia , Tolerância Imunológica/imunologia , Inflamação , Interleucina-2/imunologia , Fígado/citologia , Fígado/imunologia , Tecido Linfoide/citologia , Tecido Linfoide/imunologia , Fragmentos de Peptídeos/imunologia , Células T Auxiliares Foliculares/citologia , Células T Auxiliares Foliculares/imunologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Células Th1/citologia , Células Th1/imunologia
9.
10.
Nature ; 565(7740): 480-484, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30651642

RESUMO

The embryonic gut tube is a cylindrical structure from which the respiratory and gastrointestinal tracts develop1. Although the early emergence of the endoderm as an epithelial sheet2,3 and later morphogenesis of the definitive digestive and respiratory organs4-6 have been investigated, the intervening process of gut tube formation remains relatively understudied7,8. Here we investigate the molecular control of macroscopic forces underlying early morphogenesis of the gut tube in the chick embryo. The gut tube has been described as forming from two endodermal invaginations-the anterior intestinal portal (AIP) towards the rostral end of the embryo and the caudal intestinal portal (CIP) at the caudal end-that migrate towards one another, internalizing the endoderm until they meet at the yolk stalk (umbilicus in mammals)1,6. Migration of the AIP to form foregut has been descriptively characterized8,9, but the hindgut is likely to form by a distinct mechanism that has not been fully explained10. We find that the hindgut is formed by collective cell movements through a stationary CIP, rather than by movement of the CIP itself. Further, combining in vivo imaging, biophysics and mathematical modelling with molecular and embryological approaches, we identify a contractile force gradient that drives cell movements in the hindgut-forming endoderm, enabling tissue-scale posterior extension of the forming hindgut tube. The force gradient, in turn, is established in response to a morphogenic gradient of fibroblast growth factor signalling. As a result, we propose that an important positive feedback arises, whereby contracting cells draw passive cells from low to high fibroblast growth factor levels, recruiting them to contract and pull more cells into the elongating hindgut. In addition to providing insight into the early gut development, these findings illustrate how large-scale tissue level forces can be traced to developmental signals during vertebrate morphogenesis.


Assuntos
Trato Gastrointestinal/embriologia , Morfogênese , Animais , Padronização Corporal , Movimento Celular , Embrião de Galinha , Endoderma/citologia , Endoderma/embriologia , Endoderma/metabolismo , Fator 8 de Crescimento de Fibroblasto/metabolismo , Trato Gastrointestinal/citologia , Trato Gastrointestinal/metabolismo , Transdução de Sinais
11.
Genomics ; 116(5): 110898, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39047877

RESUMO

Using the 10x Genomics Chromium single-cell RNA sequencing (scRNA-seq) platform, we discovered unexpected heterogeneity in an established cell line developed from the midgut of the Fall armyworm, Spodoptera frugiperda, a major global pest. We analyzed the sequences of 18,794 cells and identified ten unique cellular clusters, including stem cells, enteroblasts, enterocytes and enteroendocrine cells, characterized by the expression of specific marker genes. Additionally, these studies addressed an important knowledge gap by investigating the expression of genes coding for respiratory and midgut membrane insecticide targets classified by the Insecticide Resistance Action Committee. Dual-fluorescence tagging method, fluorescence microscopy and fluorescence-activated cell sorting confirmed the expression of midgut cell type-specific genes. Stem cells were isolated from the heterogeneous population of SfMG-0617 cells. Our results, validated by KEGG and Gene Ontology analyses and supported by Monocle 3.0, advance the fields of midgut cellular biology and establish standards for scRNA-seq studies in non-model organisms.


Assuntos
Análise de Célula Única , Spodoptera , Animais , Spodoptera/genética , Spodoptera/citologia , Análise de Célula Única/métodos , Linhagem Celular , Análise de Sequência de RNA , RNA-Seq , Sistema Digestório/metabolismo , Sistema Digestório/citologia , Transcriptoma , Células-Tronco/metabolismo , Células-Tronco/citologia , Trato Gastrointestinal/citologia , Trato Gastrointestinal/metabolismo
12.
Immunity ; 41(1): 11-3, 2014 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-25035949

RESUMO

We host a world inside, and every day, new evidence reveals how relevant our microbiota is for daily living. In the most recent issue of Cell, Muller and colleagues demonstrate that microbiota commensals also influence colon peristalsis via a direct effect of muscolaris externae macrophages (Muller et al., 2014).


Assuntos
Motilidade Gastrointestinal , Trato Gastrointestinal/citologia , Trato Gastrointestinal/microbiologia , Macrófagos/metabolismo , Animais
13.
Nature ; 549(7671): 282-286, 2017 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-28869965

RESUMO

The type 2 cytokines interleukin (IL)-4, IL-5, IL-9 and IL-13 have important roles in stimulating innate and adaptive immune responses that are required for resistance to helminth infection, promotion of allergic inflammation, metabolic homeostasis and tissue repair. Group 2 innate lymphoid cells (ILC2s) produce type 2 cytokines, and although advances have been made in understanding the cytokine milieu that promotes ILC2 responses, how ILC2 responses are regulated by other stimuli remains poorly understood. Here we demonstrate that ILC2s in the mouse gastrointestinal tract co-localize with cholinergic neurons that express the neuropeptide neuromedin U (NMU). In contrast to other haematopoietic cells, ILC2s selectively express the NMU receptor 1 (NMUR1). In vitro stimulation of ILC2s with NMU induced rapid cell activation, proliferation, and secretion of the type 2 cytokines IL-5, IL-9 and IL-13 that was dependent on cell-intrinsic expression of NMUR1 and Gαq protein. In vivo administration of NMU triggered potent type 2 cytokine responses characterized by ILC2 activation, proliferation and eosinophil recruitment that was associated with accelerated expulsion of the gastrointestinal nematode Nippostrongylus brasiliensis or induction of lung inflammation. Conversely, worm burden was higher in Nmur1-/- mice than in control mice. Furthermore, use of gene-deficient mice and adoptive cell transfer experiments revealed that ILC2s were necessary and sufficient to mount NMU-elicited type 2 cytokine responses. Together, these data indicate that the NMU-NMUR1 neuronal signalling circuit provides a selective mechanism through which the enteric nervous system and innate immune system integrate to promote rapid type 2 cytokine responses that can induce anti-microbial, inflammatory and tissue-protective type 2 responses at mucosal sites.


Assuntos
Citocinas/imunologia , Imunidade Inata , Inflamação/imunologia , Linfócitos/imunologia , Neuropeptídeos/metabolismo , Transferência Adotiva , Animais , Neurônios Colinérgicos/efeitos dos fármacos , Neurônios Colinérgicos/metabolismo , Citocinas/metabolismo , Eosinófilos/citologia , Eosinófilos/efeitos dos fármacos , Eosinófilos/imunologia , Feminino , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Trato Gastrointestinal/citologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/inervação , Imunidade Inata/efeitos dos fármacos , Inflamação/induzido quimicamente , Inflamação/patologia , Interleucina-13/imunologia , Interleucina-13/metabolismo , Interleucina-5/imunologia , Interleucina-5/metabolismo , Interleucina-9/imunologia , Interleucina-9/metabolismo , Linfócitos/citologia , Linfócitos/efeitos dos fármacos , Masculino , Camundongos , Neuropeptídeos/farmacologia , Nippostrongylus/imunologia , Pneumonia/induzido quimicamente , Pneumonia/imunologia , Pneumonia/patologia , Receptores de Neurotransmissores/deficiência , Receptores de Neurotransmissores/genética , Receptores de Neurotransmissores/metabolismo , Transdução de Sinais/efeitos dos fármacos
14.
Nat Mater ; 20(1): 22-29, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32958879

RESUMO

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.


Assuntos
Bioimpressão/métodos , Organoides/metabolismo , Matriz Extracelular/metabolismo , Trato Gastrointestinal/citologia
15.
Dev Biol ; 457(1): 9-12, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31550481

RESUMO

We describe a new methodology for genetically labeling single cell lineages in Drosophila called DMARCM. The system offers ultra-low frequency labeling, linear induction, consistent labeling among individuals and virtually no background signal. We compare this technique to an existing approach, which has been widely adopted. We demonstrate how application of DMARCM in the gastrointestinal epithelium permits the effects of labeling frequency on tumorigenic stem cell growth to be distinguished in an established tumor model.


Assuntos
Linhagem da Célula , Técnicas Citológicas/métodos , Drosophila/citologia , Animais , DNA Nucleotidiltransferases , Drosophila/crescimento & desenvolvimento , Trato Gastrointestinal/citologia , Resposta ao Choque Térmico , Microscopia Confocal , Coloração e Rotulagem/métodos
16.
Physiol Rev ; 94(3): 859-907, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24987007

RESUMO

Smooth muscles are complex tissues containing a variety of cells in addition to muscle cells. Interstitial cells of mesenchymal origin interact with and form electrical connectivity with smooth muscle cells in many organs, and these cells provide important regulatory functions. For example, in the gastrointestinal tract, interstitial cells of Cajal (ICC) and PDGFRα(+) cells have been described, in detail, and represent distinct classes of cells with unique ultrastructure, molecular phenotypes, and functions. Smooth muscle cells are electrically coupled to ICC and PDGFRα(+) cells, forming an integrated unit called the SIP syncytium. SIP cells express a variety of receptors and ion channels, and conductance changes in any type of SIP cell affect the excitability and responses of the syncytium. SIP cells are known to provide pacemaker activity, propagation pathways for slow waves, transduction of inputs from motor neurons, and mechanosensitivity. Loss of interstitial cells has been associated with motor disorders of the gut. Interstitial cells are also found in a variety of other smooth muscles; however, in most cases, the physiological and pathophysiological roles for these cells have not been clearly defined. This review describes structural, functional, and molecular features of interstitial cells and discusses their contributions in determining the behaviors of smooth muscle tissues.


Assuntos
Células Intersticiais de Cajal/fisiologia , Músculo Liso/fisiologia , Animais , Trato Gastrointestinal/citologia , Trato Gastrointestinal/fisiologia , Genitália/citologia , Genitália/fisiologia , Humanos , Músculo Liso/citologia , Sistema Urinário/citologia , Fenômenos Fisiológicos do Sistema Urinário
17.
Gastroenterology ; 159(2): 453-466.e1, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32417404

RESUMO

Single cells are the building blocks of tissue systems that determine organ phenotypes, behaviors, and functions. Understanding the differences between cell types and their activities might provide us with insights into normal tissue physiology, development of disease, and new therapeutic strategies. Although -omic level single-cell technologies are a relatively recent development that have been used only in research settings, these approaches might eventually be used in the clinic. We review the prospects of applying single-cell genome, transcriptome, epigenome, proteome, and metabolome analyses to gastroenterology and hepatology research. Combining data from multi-omic platforms coupled to rapid technological development could lead to new diagnostic, prognostic, and therapeutic approaches.


Assuntos
Pesquisa Biomédica/métodos , Gastroenteropatias/diagnóstico , Trato Gastrointestinal/fisiologia , Análise de Célula Única , Gastroenteropatias/etiologia , Gastroenteropatias/fisiopatologia , Gastroenteropatias/terapia , Trato Gastrointestinal/citologia , Perfilação da Expressão Gênica/métodos , Genômica/métodos , Humanos , Metabolômica/métodos , Proteômica/métodos
18.
J Med Virol ; 93(12): 6671-6685, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34324210

RESUMO

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.


Assuntos
Enzima de Conversão de Angiotensina 2/metabolismo , Trato Gastrointestinal/virologia , Genitália/virologia , Receptor Celular 1 do Vírus da Hepatite A/metabolismo , Sistema Imunitário/virologia , Neuropilina-1/metabolismo , Sistema Respiratório/virologia , SARS-CoV-2/fisiologia , Serina Endopeptidases/metabolismo , Internalização do Vírus , Western Blotting , COVID-19/metabolismo , COVID-19/virologia , Linhagem Celular , Células Cultivadas , Imunofluorescência , Trato Gastrointestinal/citologia , Genitália/citologia , Humanos , Sistema Imunitário/citologia , Sistema Respiratório/citologia
19.
J Dairy Sci ; 104(1): 1175-1182, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33162086

RESUMO

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.


Assuntos
Antígenos de Diferenciação/metabolismo , Biomarcadores/metabolismo , Bovinos/metabolismo , Trato Gastrointestinal/metabolismo , Mucosa Intestinal/metabolismo , RNA/metabolismo , Animais , Bovinos/anatomia & histologia , Ceco/metabolismo , Células Epiteliais/metabolismo , Fezes , Trato Gastrointestinal/citologia , Íleo/metabolismo , Mucosa Intestinal/citologia , Intestino Grosso , Jejuno/metabolismo , Masculino , Reação em Cadeia da Polimerase em Tempo Real , Rúmen/metabolismo , Junções Íntimas , Transcriptoma
20.
Int J Mol Sci ; 22(5)2021 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-33652988

RESUMO

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.


Assuntos
COVID-19/patologia , Organoides/virologia , SARS-CoV-2/fisiologia , Células-Tronco/virologia , Animais , Apoptose , COVID-19/virologia , Sistema Cardiovascular/citologia , Sistema Cardiovascular/patologia , Sistema Cardiovascular/virologia , Sistema Nervoso Central/citologia , Sistema Nervoso Central/patologia , Sistema Nervoso Central/virologia , Trato Gastrointestinal/citologia , Trato Gastrointestinal/patologia , Trato Gastrointestinal/virologia , Humanos , Inflamação/patologia , Inflamação/virologia , Pulmão/citologia , Pulmão/patologia , Pulmão/virologia , Organoides/patologia , Células-Tronco/patologia , Tropismo Viral , Internalização do Vírus
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa