Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.483
Filtrar
1.
Cells ; 13(19)2024 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-39404387

RESUMO

In the small intestine, sodium (Na) absorption occurs primarily via two apical transporters, Na-hydrogen exchanger 3 (NHE3) and Na-glucose cotransporter 1 (SGLT1). The two primary Na-absorptive pathways were previously shown to compensatorily regulate each other in rabbit and rat intestinal epithelial cells. However, whether NHE3 and SGLT1 regulate one another in normal human enterocytes is unknown, mainly due to a lack of appropriate experimental models. To investigate this, we generated 2D enterocyte monolayers from human jejunal 3D organoids and used small interfering RNAs (siRNAs) to knock down NHE3 or SGLT1. Molecular and uptake studies were performed to determine the effects on NHE3 and SGLT1 expression and activity. Knockdown of NHE3 by siRNA in enterocyte monolayers was verified by qPCR and Western blot analysis and resulted in reduced NHE3 activity. However, in NHE3 siRNA-transfected cells, SGLT1 activity was significantly increased. siRNA knockdown of SGLT1 was confirmed by qPCR and Western blot analysis and resulted in reduced SGLT1 activity. However, in SGLT1 siRNA-transfected cells, NHE3 activity was significantly increased. These results demonstrate for the first time the functionality of siRNA in patient-derived organoid monolayers. Furthermore, they show that the two primary Na absorptive pathways in human enterocytes reciprocally regulate one another.


Assuntos
Enterócitos , Microvilosidades , Organoides , Transportador 1 de Glucose-Sódio , Trocador 3 de Sódio-Hidrogênio , Sódio , Humanos , Enterócitos/metabolismo , Enterócitos/citologia , Transportador 1 de Glucose-Sódio/metabolismo , Transportador 1 de Glucose-Sódio/genética , Microvilosidades/metabolismo , Trocador 3 de Sódio-Hidrogênio/metabolismo , Trocador 3 de Sódio-Hidrogênio/genética , Organoides/metabolismo , Sódio/metabolismo , RNA Interferente Pequeno/metabolismo , Jejuno/metabolismo , Jejuno/citologia , Trocadores de Sódio-Hidrogênio/metabolismo , Trocadores de Sódio-Hidrogênio/genética
2.
Cell Rep ; 43(9): 114693, 2024 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-39235946

RESUMO

Nutrient digestion, absorption, and export must be coordinated in the gut to meet the nutritional needs of the organism. We used the Drosophila intestine to characterize the mechanisms that coordinate the fate of dietary lipids. We identified enterocytes specialized in absorbing and exporting lipids to peripheral organs. Distinct hepatocyte-like cells, called oenocytes, communicate with these enterocytes to adjust intestinal lipid storage and export. A single transcription factor, Drosophila hepatocyte nuclear factor 4 (dHNF4), supports this gut-liver axis. In enterocytes, dHNF4 maximizes dietary lipid export by preventing their sequestration in cytoplasmic lipid droplets. In oenocytes, dHNF4 promotes the expression of the insulin antagonist ImpL2 to activate Foxo and suppress lipid retention in enterocytes. Disruption of this switch between lipid storage and export is associated with intestinal inflammation, suggesting a lipidic origin for inflammatory bowel diseases. These studies establish dHNF4 as a central regulator of intestinal metabolism and inter-organ lipid trafficking.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster , Enterócitos , Fator 4 Nuclear de Hepatócito , Metabolismo dos Lipídeos , Animais , Fator 4 Nuclear de Hepatócito/metabolismo , Fator 4 Nuclear de Hepatócito/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Enterócitos/metabolismo , Drosophila melanogaster/metabolismo , Mucosa Intestinal/metabolismo , Intestinos
3.
Cell Metab ; 36(10): 2341-2354.e6, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39317186

RESUMO

The exponential rise in metabolic dysfunction-associated steatotic liver disease (MASLD) parallels the ever-increasing consumption of energy-dense diets, underscoring the need for effective MASLD-resolving drugs. MASLD pathogenesis is linked to obesity, diabetes, "gut-liver axis" alterations, and defective interleukin-22 (IL-22) signaling. Although barrier-protective IL-22 blunts diet-induced metabolic alterations, inhibits lipid intake, and reverses microbial dysbiosis, obesogenic diets rapidly suppress its production by small intestine-localized innate lymphocytes. This results in STAT3 inhibition in intestinal epithelial cells (IECs) and expansion of the absorptive enterocyte compartment. These MASLD-sustaining aberrations were reversed by administration of recombinant IL-22, which resolved hepatosteatosis, inflammation, fibrosis, and insulin resistance. Exogenous IL-22 exerted its therapeutic effects through its IEC receptor, rather than hepatocytes, activating STAT3 and inhibiting WNT-ß-catenin signaling to shrink the absorptive enterocyte compartment. By reversing diet-reinforced macronutrient absorption, the main source of liver lipids, IL-22 signaling restoration represents a potentially effective interception of dietary obesity and MASLD.


Assuntos
Enterócitos , Interleucina 22 , Fator de Transcrição STAT3 , Animais , Humanos , Masculino , Camundongos , Dieta , Dieta Hiperlipídica/efeitos adversos , Enterócitos/metabolismo , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Homeostase , Interleucina 22/metabolismo , Interleucinas/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/patologia , Intestinos/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Fator de Transcrição STAT3/metabolismo
4.
Adv Exp Med Biol ; 1460: 73-95, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39287849

RESUMO

In this chapter, intestinal lipid transport, which plays a central role in fat homeostasis and the development of obesity in addition to the mechanisms of fatty acids and monoacylglycerol absorption in the intestinal lumen and reassembly of these within the enterocyte was described. A part of the resynthesized triglycerides (triacylglycerols; TAG) is repackaged in the intestine to form the hydrophobic core of chylomicrons (CMs). These are delivered as metabolic fuels, essential fatty acids, and other lipid-soluble nutrients, from enterocytes to the peripheral tissues following detachment from the endoplasmic reticulum membrane. Moreover, the attitudes of multiple receptor functions in dietary lipid uptake, synthesis, and transport are highlighted. Additionally, intestinal fatty acid binding proteins (FABPs), which increase the cytosolic flux of fatty acids via intermembrane transfer in enterocytes, and the functions of checkpoints for receptor-mediated fatty acid signaling are debated. The importance of the balance between storage and secretion of dietary fat by enterocytes in determining the physiological fate of dietary fat, including regulation of blood lipid concentrations and energy balance, is mentioned. Consequently, promising checkpoints regarding how intestinal fat processing affects lipid homeostatic mechanisms and lipid stores in the body and the prevention of obesity-lipotoxicity due to excessive intestinal lipid absorption are evaluated. In this context, dietary TAG digestion, pharmacological inhibition of TAG hydrolysis, the regulation of long-chain fatty acid uptake traffic into adipocytes, intracellular TAG resynthesis, the enlargement of cytoplasmic lipid droplets in enterocytes and constitutional alteration of their proteome, CD36-mediated conversion of diet-derived fatty acid into cellular lipid messengers and their functions are discussed.


Assuntos
Absorção Intestinal , Obesidade , Humanos , Obesidade/metabolismo , Animais , Gorduras na Dieta/metabolismo , Gorduras na Dieta/efeitos adversos , Metabolismo dos Lipídeos , Enterócitos/metabolismo , Triglicerídeos/metabolismo , Ácidos Graxos/metabolismo , Proteínas de Ligação a Ácido Graxo/metabolismo
5.
J Virol ; 98(9): e0054024, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39162433

RESUMO

Systemic viral infection of insects typically begins with the primary infection of midgut epithelial cells (enterocytes) and subsequent transit of the progeny virus in an apical-to-basal orientation into the hemocoel. For insect-vectored viruses, an oppositely oriented process (basal-to-apical transit) occurs upon secondary infection of salivary glands and is necessary for virus transmission to non-insect hosts. To examine this inversely oriented virus transit in these polarized tissues, we assessed the intracellular trafficking of two model viral envelope proteins (baculovirus GP64 and vesicular stomatitis virus G) in the midgut and salivary gland cells of the model insect, Drosophila melanogaster. Using fly lines that inducibly express either GP64 or VSV G, we found that each protein, expressed alone, was trafficked basally in midgut enterocytes. In salivary gland cells, VSV G was trafficked apically in most but not all cells, whereas GP64 was consistently trafficked basally. We demonstrated that a YxxØ motif present in both proteins was critical for basal trafficking in midgut enterocytes but dispensable for trafficking in salivary gland cells. Using RNAi, we found that clathrin adaptor protein complexes AP-1 and AP-3, as well as seven Rab GTPases, were involved in polarized VSV G trafficking in midgut enterocytes. Our results indicate that these viral envelope proteins encode the requisite information and require no other viral factors for appropriately polarized trafficking. In addition, they exploit tissue-specific differences in protein trafficking pathways to facilitate virus egress in the appropriate orientation for establishing systemic infections and vectoring infection to other hosts. IMPORTANCE: Viruses that use insects as hosts must navigate specific routes through different insect tissues to complete their life cycles. The routes may differ substantially depending on the life cycle of the virus. Both insect pathogenic viruses and insect-vectored viruses must navigate through the polarized cells of the midgut epithelium to establish a systemic infection. In addition, insect-vectored viruses must also navigate through the polarized salivary gland epithelium for transmission. Thus, insect-vectored viruses appear to traffic in opposite directions in these two tissues. In this study, we asked whether two viral envelope proteins (VSV G and baculovirus GP64) alone encode the signals necessary for the polarized trafficking associated with their respective life cycles. Using Drosophila as a model to examine tissue-specific polarized trafficking of these viral envelope proteins, we identified one of the virus-encoded signals and several host proteins associated with regulating the polarized trafficking in the midgut epithelium.


Assuntos
Drosophila melanogaster , Transporte Proteico , Glândulas Salivares , Proteínas do Envelope Viral , Animais , Glândulas Salivares/virologia , Glândulas Salivares/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/genética , Drosophila melanogaster/virologia , Drosophila melanogaster/metabolismo , Insetos Vetores/virologia , Insetos Vetores/metabolismo , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Enterócitos/virologia , Enterócitos/metabolismo , Trato Gastrointestinal/virologia , Trato Gastrointestinal/metabolismo
6.
J Exp Med ; 221(10)2024 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-39150450

RESUMO

Intestinal stem cells at the crypt divide and give rise to progenitor cells that proliferate and differentiate into various mature cell types in the transit-amplifying (TA) zone. Here, we showed that the transcription factor ARID3A regulates intestinal epithelial cell proliferation and differentiation at the TA progenitors. ARID3A forms an expression gradient from the villus tip to the upper crypt mediated by TGF-ß and WNT. Intestinal-specific deletion of Arid3a reduces crypt proliferation, predominantly in TA cells. Bulk and single-cell transcriptomic analysis shows increased enterocyte and reduced secretory differentiation in the Arid3a cKO intestine, accompanied by enriched upper-villus gene signatures of both cell lineages. We find that the enhanced epithelial differentiation in the Arid3a-deficient intestine is caused by increased binding and transcription of HNF1 and HNF4. Finally, we show that loss of Arid3a impairs irradiation-induced regeneration with sustained cell death and reprogramming. Our findings imply that Arid3a functions to fine-tune the proliferation-differentiation dynamics at the TA progenitors, which are essential for injury-induced regeneration.


Assuntos
Diferenciação Celular , Proliferação de Células , Proteínas de Ligação a DNA , Fator 1-alfa Nuclear de Hepatócito , Mucosa Intestinal , Camundongos Knockout , Regeneração , Fatores de Transcrição , Animais , Mucosa Intestinal/metabolismo , Mucosa Intestinal/citologia , Camundongos , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/deficiência , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Fator 1-alfa Nuclear de Hepatócito/genética , Fator 4 Nuclear de Hepatócito/metabolismo , Fator 4 Nuclear de Hepatócito/genética , Células-Tronco/metabolismo , Células-Tronco/citologia , Camundongos Endogâmicos C57BL , Fator de Crescimento Transformador beta/metabolismo , Células Epiteliais/metabolismo , Enterócitos/metabolismo , Enterócitos/citologia
7.
J Microbiol ; 62(9): 799-809, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39212866

RESUMO

Dengue virus (DENV), from the Flaviviridae family, is the causative agent of dengue fever and poses a significant global health challenge. The virus primarily affects the vascular system and liver; however, a growing body of evidence suggests its involvement in the gastrointestinal (GI) tract, contributing to clinical symptoms such as abdominal pain, vomiting, and diarrhea. However, the mechanisms underlying DENV infection in the digestive system remain largely unexplored. Prior research has detected viral RNA in the GI tissue of infected animals; however, whether the dengue virus can directly infect human enterocytes remains unclear. In this study, we examine the infectivity of human intestinal cell lines to the dengue virus and their subsequent response. We report that the Caco-2 cell line, a model of human enterocytes, is susceptible to infection and capable of producing viruses. Notably, differentiated Caco-2 cells exhibited a lower infection rate yet a higher level of virus production than their undifferentiated counterparts. These findings suggest that human intestinal cells are a viable target for the dengue virus, potentially elucidating the GI symptoms observed in dengue fever and offering a new perspective on the pathogenetic mechanisms of the virus.


Assuntos
Diferenciação Celular , Vírus da Dengue , Dengue , Enterócitos , Humanos , Células CACO-2 , Vírus da Dengue/fisiologia , Vírus da Dengue/patogenicidade , Enterócitos/virologia , Dengue/virologia , Replicação Viral , RNA Viral/genética
8.
Nature ; 632(8027): 1101-1109, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39112711

RESUMO

The mouse small intestine shows profound variability in gene expression along the crypt-villus axis1,2. Whether similar spatial heterogeneity exists in the adult human gut remains unclear. Here we use spatial transcriptomics, spatial proteomics and single-molecule fluorescence in situ hybridization to reconstruct a comprehensive spatial expression atlas of the adult human proximal small intestine. We describe zonated expression and cell type representation for epithelial, mesenchymal and immune cell types. We find that migrating enterocytes switch from lipid droplet assembly and iron uptake at the villus bottom to chylomicron biosynthesis and iron release at the tip. Villus tip cells are pro-immunogenic, recruiting γδ T cells and macrophages to the tip, in contrast to their immunosuppressive roles in mouse. We also show that the human small intestine contains abundant serrated and branched villi that are enriched at the tops of circular folds. Our study presents a detailed resource for understanding the biology of the adult human small intestine.


Assuntos
Biologia Celular , Perfilação da Expressão Gênica , Intestino Delgado , Adulto , Animais , Feminino , Humanos , Masculino , Camundongos , Movimento Celular , Quilomícrons/biossíntese , Enterócitos/metabolismo , Enterócitos/citologia , Células Epiteliais , Hibridização in Situ Fluorescente , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Intestino Delgado/citologia , Intestino Delgado/imunologia , Intestino Delgado/metabolismo , Ferro/metabolismo , Gotículas Lipídicas/metabolismo , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , Proteômica , Imagem Individual de Molécula , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Transcriptoma
9.
Microbiol Spectr ; 12(10): e0030424, 2024 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-39189752

RESUMO

Atypical enteropathogenic Escherichia coli (aEPEC) is a significant cause of diarrhea in low- and middle-income countries. Certain aEPEC strains, including the Brazilian representative strain of serotype O51:H40 called aEPEC 1711-4, can use flagella to attach to, invade, and persist in T84 and Caco-2 intestinal cells. It can also translocate from the gut to extraintestinal sites in a rat model. Although various aspects of the virulence of this strain were studied and the requirement of a type III secretion system for the efficiency of the invasion process was demonstrated, the expression of the locus of enterocyte effacement (LEE) genes during the invasion and intracellular persistence remains unclear. To address this question, the expression of flagella and the different LEE operons was evaluated during kinetic experiments of the interaction of aEPEC 1711-4 with enterocytes in vitro. The genome of the strain was also sequenced. The results showed that flagella expression remained unchanged, but the expression of eae and escJ increased during the early interaction and invasion of aEPEC 1711-4 into Caco-2 cells, and there was no change 24 h post-infection during the persistence period. The number of actin accumulation foci formed on HeLa cells also increased during the 6-h analysis. No known gene related to the invasion process was identified in the genome of aEPEC 1711-4, which was shown to belong to the global EPEC lineage 10. These findings suggest that the LEE components and the intimate adherence promoted by intimin are necessary for the invasion and persistence of aEPEC 1711-4, but the detailed mechanism needs further study.IMPORTANCEAtypical enteropathogenic Escherichia coli (aEPEC) is a major cause of diarrhea, especially in low- and middle-income countries, like Brazil. However, due to the genome heterogeneity of each clonal group, it is difficult to comprehend the pathogenicity of this strain fully. Among aEPEC strains, 1711-4 can invade eukaryotic cells in vitro, cross the gut barrier, and reach extraintestinal sites in animal models. By studying how different known aEPEC virulence factors are expressed during the invasion process, we can gain insight into the commonalities of this phenotype among other aEPEC strains. This will help in developing preventive measures to control infections caused by invasive strains. No known virulence-encoding genes linked to the invasion process were found. Nevertheless, additional studies are still necessary to evaluate the role of other factors in this phenotype.


Assuntos
Enterócitos , Escherichia coli Enteropatogênica , Infecções por Escherichia coli , Proteínas de Escherichia coli , Flagelos , Sorogrupo , Escherichia coli Enteropatogênica/genética , Escherichia coli Enteropatogênica/patogenicidade , Escherichia coli Enteropatogênica/metabolismo , Humanos , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Enterócitos/microbiologia , Células CACO-2 , Infecções por Escherichia coli/microbiologia , Flagelos/genética , Flagelos/metabolismo , Virulência/genética , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Regulação Bacteriana da Expressão Gênica , Aderência Bacteriana/genética , Animais , Brasil , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Óperon/genética , Ratos
10.
FASEB J ; 38(15): e23847, 2024 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-39096137

RESUMO

Intestinal failure-associated liver disease (IFALD) is a serious complication of long-term parenteral nutrition in patients with short bowel syndrome (SBS), and is the main cause of death in SBS patients. Prevention of IFALD is one of the major challenges in the treatment of SBS. Impairment of intestinal barrier function is a key factor in triggering IFALD, therefore promoting intestinal repair is particularly important. Intestinal repair mainly relies on the function of intestinal stem cells (ISC), which require robust mitochondrial fatty acid oxidation (FAO) for self-renewal. Herein, we report that aberrant LGR5+ ISC function in IFALD may be attributed to impaired farnesoid X receptor (FXR) signaling, a transcriptional factor activated by steroids and bile acids. In both surgical biopsies and patient-derived organoids (PDOs), SBS patients with IFALD represented lower population of LGR5+ cells and decreased FXR expression. Moreover, treatment with T-ßMCA in PDOs (an antagonist for FXR) dose-dependently reduced the population of LGR5+ cells and the proliferation rate of enterocytes, concomitant with decreased key genes involved in FAO including CPT1a. Interestingly, however, treatment with Tropifexor in PDOs (an agonist for FXR) only enhanced FAO capacity, without improvement in ISC function and enterocyte proliferation. In conclusion, these findings suggested that impaired FXR may accelerate the depletion of LGR5 + ISC population through disrupted FAO processes, which may serve as a new potential target of preventive interventions against IFALD for SBS patients.


Assuntos
Hepatopatias , Receptores Citoplasmáticos e Nucleares , Síndrome do Intestino Curto , Transdução de Sinais , Células-Tronco , Humanos , Síndrome do Intestino Curto/metabolismo , Síndrome do Intestino Curto/patologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Células-Tronco/metabolismo , Masculino , Hepatopatias/metabolismo , Hepatopatias/patologia , Hepatopatias/etiologia , Feminino , Criança , Insuficiência Intestinal/metabolismo , Pré-Escolar , Lactente , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Receptores Acoplados a Proteínas G/metabolismo , Proliferação de Células , Intestinos/patologia , Enterócitos/metabolismo
11.
Nat Microbiol ; 9(9): 2448-2461, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38965331

RESUMO

Interactions between microbiota and enteric pathogens can promote colonization resistance or enhance pathogenesis. The pathobiont Enterococcus faecalis increases enterohaemorrhagic E. coli (EHEC) virulence by upregulating Type 3 Secretion System (T3SS) expression, effector translocation, and attaching and effacing (AE) lesion formation on enterocytes, but the mechanisms underlying this remain unknown. Using co-infection of organoids, metabolomics, supplementation experiments and bacterial genetics, here we show that co-culture of EHEC with E. faecalis increases the xanthine-hypoxanthine pathway activity and adenine biosynthesis. Adenine or E. faecalis promoted T3SS gene expression, while transcriptomics showed upregulation of adeP expression, which encodes an adenine importer. Mechanistically, adenine relieved High hemolysin activity (Hha)-dependent repression of T3SS gene expression in EHEC and promoted AE lesion formation in an AdeP-dependent manner. Microbiota-derived purines, such as adenine, support multiple beneficial host responses; however, our data show that this metabolite also increases EHEC virulence, highlighting the complexity of pathogen-microbiota-host interactions in the gut.


Assuntos
Adenina , Enterococcus faecalis , Escherichia coli Êntero-Hemorrágica , Regulação Bacteriana da Expressão Gênica , Sistemas de Secreção Tipo III , Escherichia coli Êntero-Hemorrágica/genética , Escherichia coli Êntero-Hemorrágica/patogenicidade , Escherichia coli Êntero-Hemorrágica/metabolismo , Virulência , Sistemas de Secreção Tipo III/metabolismo , Sistemas de Secreção Tipo III/genética , Enterococcus faecalis/genética , Enterococcus faecalis/metabolismo , Enterococcus faecalis/patogenicidade , Adenina/metabolismo , Adenina/farmacologia , Animais , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Camundongos , Infecções por Escherichia coli/microbiologia , Humanos , Proteínas Hemolisinas/metabolismo , Proteínas Hemolisinas/genética , Interações Hospedeiro-Patógeno , Técnicas de Cocultura , Enterócitos/microbiologia , Enterócitos/metabolismo , Xantina/metabolismo , Hipoxantina/metabolismo , Fatores de Virulência/metabolismo , Fatores de Virulência/genética , Microbioma Gastrointestinal
12.
Endocrinology ; 165(8)2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38953181

RESUMO

Neprilysin is a ubiquitous peptidase that can modulate glucose homeostasis by cleaving insulinotropic peptides. While global deletion of neprilysin protects mice against high-fat diet (HFD)-induced insulin secretory dysfunction, strategies to ablate neprilysin in a tissue-specific manner are favored to limit off-target effects. Since insulinotropic peptides are produced in the gut, we sought to determine whether gut-specific neprilysin deletion confers beneficial effects on insulin secretion similar to that of global neprilysin deletion in mice fed a HFD. Mice with conditional deletion of neprilysin in enterocytes (NEPGut-/-) were generated by crossing Vil-Cre and floxed neprilysin mice. Neprilysin activity was almost abolished throughout the gut in NEPGut-/- mice, and was similar in plasma, pancreas, and kidney in NEPGut-/- vs control mice. An oral glucose tolerance test was performed at baseline and following 14 weeks of HFD feeding, during which glucose tolerance and glucose-stimulated insulin secretion (GSIS) were assessed. Despite similar body weight gain at 14 weeks, NEPGut-/- displayed lower fasting plasma glucose levels, improved glucose tolerance, and increased GSIS compared to control mice. In conclusion, gut-specific neprilysin deletion recapitulates the enhanced GSIS seen with global neprilysin deletion in HFD-fed mice. Thus, strategies to inhibit neprilysin specifically in the gut may protect against fat-induced glucose intolerance and beta-cell dysfunction.


Assuntos
Dieta Hiperlipídica , Secreção de Insulina , Insulina , Neprilisina , Animais , Masculino , Camundongos , Dieta Hiperlipídica/efeitos adversos , Enterócitos/metabolismo , Deleção de Genes , Teste de Tolerância a Glucose , Insulina/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neprilisina/genética , Neprilisina/metabolismo
13.
PLoS One ; 19(7): e0307411, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39024309

RESUMO

The microbiological environment and their corresponding secreted metabolite spectrum are an essential modulator of the enterocyte function, effecting the whole organism. Intestinal porcine jejunal epithelial cell line (IPEC-J2) is an established in vitro model for differentiation of enterocytes in different cell culture models. An improved oxygen supply seems to be the main reason for differentiation in an air-liquid-interface culture, but this has not yet been conclusively clarified. In this context, the nutrition of the cell and its influence on the metabolism is also of crucial importance. The interest in short-chain fatty acids (SCFAs) has grown steadily in recent years due to their clinical relevance in certain diseases such as multiple sclerosis and other inflammatory diseases, but not much is known of FFAR2 and FFAR3 (free fatty acid receptor 2 and 3) in pigs. We want to address the questions: 1. about the distribution of FFAR2 and FFAR3 in vivo and in vitro in sus scrofa 2. whether there is an influence of propionic acid, glucose content and cultivation on metabolism of enterocytes? The morphological analysis of FFAR2 and FFAR3 in vivo was investigated through immunostaining of frozen sections of the porcine gut segments jejunum, ileum and colon. Both receptors are expressed along the gut and were found in the smooth muscle cells of the tunica muscularis and lamina muscularis mucosae. Furthermore, a high expression of FFAR2 and a low expression of FFAR3 in the enteric nerve system was also observed in jejunum, ileum and colon of sus scrofa. In addition, FFAR2 and FFAR3 within the vessels was investigated. FFAR3 showed a strong expression on endothelial cells of veins and lymphatic vessels but was not detectable on arteries. Furthermore, we demonstrate for the first time, FFAR2 and FFAR3 in IPEC-J2 cells on RNA- and protein level, as well as with confocal microscopy. In addition, ENO1 and NDUFA4 were investigated on RNA-level in IPEC-J2 cells as 2 important genes, which play an essential role in metabolism. Here, NDUFA4 is detected in the model animal sus scrofa as well as in the porcine cell line IPEC-J2. A potential impact of propionic acid and/or glucose and/or cultivation method on the metabolism of the cells was tested with the Seahorse analyzer. Here, a significant higher ECAR was observed in the SMC than in the OCR. In summary, we were able to show that the cultivation system appears to have a greater influence than the medium composition or nutrition of the cells. However, this can be modulated by incubation time or combination of different SCFAs.


Assuntos
Glucose , Propionatos , Animais , Propionatos/metabolismo , Glucose/metabolismo , Suínos , Linhagem Celular , Técnicas de Cultura de Células/métodos , Metaboloma , Receptores Acoplados a Proteínas G/metabolismo , Jejuno/metabolismo , Jejuno/citologia , Enterócitos/metabolismo , Mucosa Intestinal/metabolismo
14.
EMBO J ; 43(16): 3466-3493, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38965418

RESUMO

The gut microbiota and their metabolites are closely linked to obesity-related diseases, such as type 2 diabetes, but their causal relationship and underlying mechanisms remain largely elusive. Here, we found that dysbiosis-induced tyramine (TA) suppresses high-fat diet (HFD)-mediated insulin resistance in both Drosophila and mice. In Drosophila, HFD increases cytosolic Ca2+ signaling in enterocytes, which, in turn, suppresses intestinal lipid levels. 16 S rRNA sequencing and metabolomics revealed that HFD leads to increased prevalence of tyrosine decarboxylase (Tdc)-expressing bacteria and resulting tyramine production. Tyramine acts on the tyramine receptor, TyrR1, to promote cytosolic Ca2+ signaling and activation of the CRTC-CREB complex to transcriptionally suppress dietary lipid digestion and lipogenesis in enterocytes, while promoting mitochondrial biogenesis. Furthermore, the tyramine-induced cytosolic Ca2+ signaling is sufficient to suppress HFD-induced obesity and insulin resistance in Drosophila. In mice, tyramine intake also improves glucose tolerance and insulin sensitivity under HFD. These results indicate that dysbiosis-induced tyramine suppresses insulin resistance in both flies and mice under HFD, suggesting a potential therapeutic strategy for related metabolic disorders, such as diabetes.


Assuntos
Sinalização do Cálcio , Dieta Hiperlipídica , Microbioma Gastrointestinal , Resistência à Insulina , Tiramina , Animais , Tiramina/metabolismo , Tiramina/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Dieta Hiperlipídica/efeitos adversos , Camundongos , Sinalização do Cálcio/efeitos dos fármacos , Obesidade/metabolismo , Obesidade/microbiologia , Obesidade/etiologia , Masculino , Drosophila/metabolismo , Disbiose/metabolismo , Disbiose/microbiologia , Camundongos Endogâmicos C57BL , Drosophila melanogaster/microbiologia , Drosophila melanogaster/metabolismo , Enterócitos/metabolismo , Enterócitos/efeitos dos fármacos
15.
Eur J Pharm Biopharm ; 202: 114408, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39004319

RESUMO

The therapeutic effects of orally administered nanocarriers depend on their ability to effectively permeate the intestinal mucosa, which is one of the major challenges in oral drug delivery. Microfold cells are specialized enterocytes in the intestinal epithelium known for their high transcytosis abilities. This study aimed to compare and evaluate two targeting approaches using surface modifications of polymer-based nanocarriers, whereas one generally addresses enterocytes, and one is directed explicitly to microfold cells via targeting the sialyl LewisA motif on their surface. We characterized the resulting carriers in terms of size and charge, supplemented by scanning electron microscopy to confirm their structural properties. For predictive biological testing and to assess the intended targeting effect, we implemented two human intestinal in vitro models containing microfold-like cells. Both models were thoroughly characterized prior to permeation studies with the different nanocarriers. Our results demonstrated improved transport for both targeted formulations compared to undecorated carriers in the in vitro models. Notably, there was an enhanced uptake in the presence of microfold-like cells, particularly for the nanocarriers directed by the anti-sialyl LewisA antibody. These findings highlight the potential of microfold cell targeting to improve oral administration of drugs and emphasize the importance of using suitable and well-characterized in vitro models for testing novel drug delivery strategies.


Assuntos
Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Mucosa Intestinal , Células M , Nanopartículas , Humanos , Administração Oral , Células CACO-2 , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos/métodos , Enterócitos/metabolismo , Enterócitos/efeitos dos fármacos , Absorção Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Células M/metabolismo , Nanopartículas/química , Permeabilidade , Polímeros/química
16.
mBio ; 15(8): e0131624, 2024 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-38953637

RESUMO

Human intestinal enteroids (HIEs) are gaining recognition as physiologically relevant models of the intestinal epithelium. While HIEs from adults are used extensively in biomedical research, few studies have used HIEs from infants. Considering the dramatic developmental changes that occur during infancy, it is important to establish models that represent infant intestinal characteristics and physiological responses. We established jejunal HIEs from infant surgical samples and performed comparisons to jejunal HIEs from adults using RNA sequencing (RNA-Seq) and morphologic analyses. We then validated differences in key pathways through functional studies and determined whether these cultures recapitulate known features of the infant intestinal epithelium. RNA-Seq analysis showed significant differences in the transcriptome of infant and adult HIEs, including differences in genes and pathways associated with cell differentiation and proliferation, tissue development, lipid metabolism, innate immunity, and biological adhesion. Validating these results, we observed a higher abundance of cells expressing specific enterocyte, goblet cell, and enteroendocrine cell markers in differentiated infant HIE monolayers, and greater numbers of proliferative cells in undifferentiated 3D cultures. Compared to adult HIEs, infant HIEs portray characteristics of an immature gastrointestinal epithelium including significantly shorter cell height, lower epithelial barrier integrity, and lower innate immune responses to infection with an oral poliovirus vaccine. HIEs established from infant intestinal tissues reflect characteristics of the infant gut and are distinct from adult cultures. Our data support the use of infant HIEs as an ex vivo model to advance studies of infant-specific diseases and drug discovery for this population. IMPORTANCE: Tissue or biopsy stem cell-derived human intestinal enteroids are increasingly recognized as physiologically relevant models of the human gastrointestinal epithelium. While enteroids from adults and fetal tissues have been extensively used for studying many infectious and non-infectious diseases, there are few reports on enteroids from infants. We show that infant enteroids exhibit both transcriptomic and morphological differences compared to adult cultures. They also differ in functional responses to barrier disruption and innate immune responses to infection, suggesting that infant and adult enteroids are distinct model systems. Considering the dramatic changes in body composition and physiology that begin during infancy, tools that appropriately reflect intestinal development and diseases are critical. Infant enteroids exhibit key features of the infant gastrointestinal epithelium. This study is significant in establishing infant enteroids as age-appropriate models for infant intestinal physiology, infant-specific diseases, and responses to pathogens.


Assuntos
Mucosa Intestinal , Humanos , Lactente , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Adulto , Diferenciação Celular , Jejuno/citologia , Jejuno/imunologia , Transcriptoma , Organoides , Imunidade Inata , Feminino , Masculino , Recém-Nascido , Enterócitos
17.
Am J Physiol Gastrointest Liver Physiol ; 327(3): G438-G453, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-38860856

RESUMO

Keratin intermediate filaments form dynamic filamentous networks, which provide mechanical stability, scaffolding, and protection against stress to epithelial cells. Keratins and other intermediate filaments have been increasingly linked to the regulation of mitochondrial function and homeostasis in different tissues and cell types. While deletion of keratin 8 (K8-/-) in mouse colon elicits a colitis-like phenotype, epithelial hyperproliferation, and blunted mitochondrial ketogenesis, the role of K8 in colonocyte mitochondrial function and energy metabolism is unknown. We used two K8 knockout mouse models and CRISPR/Cas9 K8-/- colorectal adenocarcinoma Caco-2 cells to answer this question. The results show that K8-/- colonocyte mitochondria in vivo are smaller and rounder and that mitochondrial motility is increased in K8-/- Caco-2 cells. Furthermore, K8-/- Caco-2 cells displayed diminished mitochondrial respiration and decreased mitochondrial membrane potential compared with controls, whereas glycolysis was not affected. The levels of mitochondrial respiratory chain complex proteins and mitochondrial regulatory proteins mitofusin-2 and prohibitin were decreased both in vitro in K8-/- Caco-2 cells and in vivo in K8-/- mouse colonocytes, and reexpression of K8 into K8-/- Caco-2 cells normalizes the mitofusin-2 levels. Mitochondrial Ca2+ is an important regulator of mitochondrial energy metabolism and homeostasis, and Caco-2 cells lacking K8 displayed decreased levels and altered dynamics of mitochondrial matrix and cytoplasmic Ca2+. In summary, these novel findings attribute an important role for colonocyte K8 in stabilizing mitochondrial shape and movement and maintaining mitochondrial respiration and Ca2+ signaling. Further, how these metabolically compromised colonocytes are capable of hyperproliferating presents an intriguing question for future studies.NEW & NOTEWORTHY In this study, we show that colonocyte intermediate filament protein keratin 8 is important for stabilizing mitochondria and maintaining mitochondrial energy metabolism, as keratin 8-deficient colonocytes display smaller, rounder, and more motile mitochondria, diminished mitochondrial respiration, and altered Ca2+ dynamics. Changes in fusion-regulating proteins are rescued with reexpression of keratin 8. These alterations in colonocyte mitochondrial homeostasis contribute to keratin 8-associated colitis pathophysiology.


Assuntos
Colo , Metabolismo Energético , Queratina-8 , Camundongos Knockout , Mitocôndrias , Animais , Mitocôndrias/metabolismo , Células CACO-2 , Humanos , Queratina-8/metabolismo , Queratina-8/genética , Colo/metabolismo , Camundongos , Proibitinas , GTP Fosfo-Hidrolases/metabolismo , GTP Fosfo-Hidrolases/genética , Proteínas Mitocondriais/metabolismo , Proteínas Mitocondriais/genética , Enterócitos/metabolismo , Potencial da Membrana Mitocondrial , Proteínas Repressoras/metabolismo , Proteínas Repressoras/genética
18.
Blood ; 144(13): 1433-1444, 2024 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-38861671

RESUMO

ABSTRACT: Ferroportin (Fpn) is the only iron exporter, playing a crucial role in systemic iron homeostasis. Fpn is negatively regulated by its ligand hepcidin, but other potential regulators in physiological and disease conditions remain poorly understood. Diabetes is a metabolic disorder that develops body iron loading with unknown mechanisms. By using diabetic mouse models and human duodenal specimens, we demonstrated that intestinal Fpn expression was increased in diabetes in a hepcidin-independent manner. Protein kinase C (PKC) is hyperactivated in diabetes. We showed that PKCα was required to sustain baseline Fpn expression and diabetes-induced Fpn upregulation in the enterocytes and macrophages. Knockout of PKCα abolished diabetes-associated iron overload. Mechanistically, activation of PKCα increased the exocytotic trafficking of Fpn and decreased the endocytic trafficking of Fpn in the resting state. Hyperactive PKCα also suppressed hepcidin-induced ubiquitination, internalization, and degradation of Fpn. We further observed that iron loading in the enterocytes and macrophages activated PKCα, acting as a novel mechanism to enhance Fpn-dependent iron efflux. Finally, we demonstrated that the loss-of-function of PKCα and pharmacological inhibition of PKC significantly alleviated hereditary hemochromatosis-associated iron overload. Our study has highlighted, to our knowledge, for the first time, that PKCα is an important positive regulator of Fpn and a new target in the control of iron homeostasis.


Assuntos
Proteínas de Transporte de Cátions , Hemocromatose , Hepcidinas , Sobrecarga de Ferro , Proteína Quinase C-alfa , Animais , Sobrecarga de Ferro/metabolismo , Proteína Quinase C-alfa/metabolismo , Proteína Quinase C-alfa/genética , Camundongos , Humanos , Proteínas de Transporte de Cátions/metabolismo , Proteínas de Transporte de Cátions/genética , Hemocromatose/metabolismo , Hemocromatose/genética , Hemocromatose/patologia , Hepcidinas/metabolismo , Hepcidinas/genética , Camundongos Knockout , Masculino , Ferro/metabolismo , Diabetes Mellitus Experimental/metabolismo , Camundongos Endogâmicos C57BL , Enterócitos/metabolismo , Enterócitos/patologia , Macrófagos/metabolismo
20.
Cell Biol Toxicol ; 40(1): 48, 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38900277

RESUMO

Aggregation of aberrant proteins is a common pathological hallmark in neurodegeneration such as polyglutamine (polyQ) and other repeat-expansion diseases. Here through overexpression of ataxin3 C-terminal polyQ expansion in Drosophila gut enterocytes, we generated an intestinal obstruction model of spinocerebellar ataxia type3 (SCA3) and reported a new role of nuclear-associated endosomes (NAEs)-the delivery of polyQ to the nucleoplasm. In this model, accompanied by the prominently increased RAB5-positive NAEs are abundant nucleoplasmic reticulum enriched with polyQ, abnormal nuclear envelope invagination, significantly reduced endoplasmic reticulum, indicating dysfunctional nucleocytoplasmic trafficking and impaired endomembrane organization. Consistently, Rab5 but not Rab7 RNAi further decreased polyQ-related NAEs, inhibited endomembrane disorganization, and alleviated disease model. Interestingly, autophagic proteins were enriched in polyQ-related NAEs and played non-canonical autophagic roles as genetic manipulation of autophagic molecules exhibited differential impacts on NAEs and SCA3 toxicity. Namely, the down-regulation of Atg1 or Atg12 mitigated while Atg5 RNAi aggravated the disease phenotypes both in Drosophila intestines and compound eyes. Our findings, therefore, provide new mechanistic insights and underscore the fundamental roles of endosome-centered nucleocytoplasmic trafficking and homeostatic endomembrane allocation in the pathogenesis of polyQ diseases.


Assuntos
Autofagia , Endossomos , Peptídeos , Animais , Peptídeos/metabolismo , Endossomos/metabolismo , Núcleo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Transporte Ativo do Núcleo Celular , Drosophila melanogaster/metabolismo , Drosophila melanogaster/genética , Doença de Machado-Joseph/metabolismo , Doença de Machado-Joseph/genética , Doença de Machado-Joseph/patologia , Enterócitos/metabolismo , Modelos Animais de Doenças , Ataxina-3/metabolismo , Ataxina-3/genética , Drosophila/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA