Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 298(3): 101594, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35041825

RESUMEN

Locally advanced rectal cancer is typically treated with chemoradiotherapy followed by surgery. Most patients do not display a complete response to chemoradiotherapy, but resistance mechanisms are poorly understood. ST6GAL-1 is a sialyltransferase that adds the negatively charged sugar, sialic acid (Sia), to cell surface proteins in the Golgi, altering their function. We therefore hypothesized that ST6GAL-1 could mediate resistance to chemoradiation in rectal cancer by inhibiting apoptosis. Patient-derived xenograft and organoid models of rectal cancer and rectal cancer cell lines were assessed for ST6GAL-1 protein with and without chemoradiation treatment. ST6GAL-1 mRNA was assessed in untreated human rectal adenocarcinoma by PCR assays. Samples were further assessed by Western blotting, Caspase-Glo apoptosis assays, and colony formation assays. The presence of functional ST6GAL-1 was assessed via flow cytometry using the Sambucus nigra lectin, which specifically binds cell surface α2,6-linked Sia, and via lectin precipitation. In patient-derived xenograft models of rectal cancer, we found that ST6GAL-1 protein was increased after chemoradiation in a subset of samples. Rectal cancer cell lines demonstrated increased ST6GAL-1 protein and cell surface Sia after chemoradiation. ST6GAL-1 was also increased in rectal cancer organoids after treatment. ST6GAL-1 knockdown in rectal cancer cell lines resulted in increased apoptosis and decreased survival after treatment. We concluded that ST6GAL-1 promotes resistance to chemoradiotherapy by inhibiting apoptosis in rectal cancer cell lines. More research will be needed to further elucidate the importance and mechanism of ST6GAL-1-mediated resistance.


Asunto(s)
Antígenos CD , Neoplasias del Recto , Sialiltransferasas , Antígenos CD/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Quimioradioterapia , Resistencia a Antineoplásicos , Humanos , Ácido N-Acetilneuramínico/metabolismo , Tolerancia a Radiación , Neoplasias del Recto/tratamiento farmacológico , Neoplasias del Recto/metabolismo , Neoplasias del Recto/patología , Neoplasias del Recto/radioterapia , Sialiltransferasas/genética , Sialiltransferasas/metabolismo , beta-D-Galactósido alfa 2-6-Sialiltransferasa
2.
J Biol Chem ; 295(41): 14153-14163, 2020 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-32763973

RESUMEN

Programmed cell death promotes homeostatic cell turnover in the epithelium but is dysregulated in cancer. The glycosyltransferase ST6Gal-I is known to block homeostatic apoptosis through α2,6-linked sialylation of the death receptor TNFR1 in many cell types. However, its role has not been investigated in gastric epithelial cells or gastric tumorigenesis. We determined that human gastric antral epithelium rarely expressed ST6Gal-I, but the number of ST6Gal-I-expressing epithelial cells increased significantly with advancing premalignancy leading to cancer. The mRNA expression levels of ST6GAL-I and SOX9 in human gastric epithelial cells correlated positively with one another through the premalignancy cascade, indicating that increased epithelial cell expression of ST6Gal-I is associated with premalignant progression. To determine the functional impact of increased ST6Gal-I, we generated human gastric antral organoids from epithelial stem cells and differentiated epithelial monolayers from gastric organoids. Gastric epithelial stem cells strongly expressed ST6Gal-I, suggesting a novel biomarker of stemness. In contrast, organoid-derived epithelial monolayers expressed markedly reduced ST6Gal-I and underwent TNF-induced, caspase-mediated apoptosis, consistent with homeostasis. Conversely, epithelial monolayers generated from gastric cancer stem cells retained high levels of ST6Gal-I and resisted TNF-induced apoptosis, supporting prolonged survival. Protection from TNF-induced apoptosis depended on ST6Gal-I overexpression, because forced ST6Gal-I overexpression in normal gastric stem cell-differentiated monolayers inhibited TNF-induced apoptosis, and cleavage of α2,6-linked sialic acids from gastric cancer organoid-derived monolayers restored susceptibility to TNF-induced apoptosis. These findings implicate up-regulated ST6Gal-I expression in blocking homeostatic epithelial cell apoptosis in gastric cancer pathogenesis, suggesting a mechanism for prolonged epithelioid tumor cell survival.


Asunto(s)
Antígenos CD/biosíntesis , Células Epiteliales/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Homeostasis , Proteínas de Neoplasias/biosíntesis , Organoides/metabolismo , Sialiltransferasas/biosíntesis , Neoplasias Gástricas/epidemiología , Antígenos CD/genética , Línea Celular , Células Epiteliales/patología , Humanos , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Organoides/patología , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Sialiltransferasas/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Células Tumorales Cultivadas
3.
J Immunol ; 193(11): 5604-12, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25355920

RESUMEN

Circulating monocytes carrying human CMV (HCMV) migrate into tissues, where they differentiate into HCMV-infected resident macrophages that upon interaction with bacterial products may potentiate tissue inflammation. In this study, we investigated the mechanism by which HCMV promotes macrophage-orchestrated inflammation using a clinical isolate of HCMV (TR) and macrophages derived from primary human monocytes. HCMV infection of the macrophages, which was associated with viral DNA replication, significantly enhanced TNF-α, IL-6, and IL-8 gene expression and protein production in response to TLR4 ligand (LPS) stimulation compared with mock-infected LPS-stimulated macrophages during a 6-d in vitro infection. HCMV infection also potentiated TLR5 ligand-stimulated cytokine production. To elucidate the mechanism by which HCMV infection potentiated inducible macrophage responses, we show that infection by HCMV promoted the maintenance of surface CD14 and TLR4 and TLR5, which declined over time in mock-infected macrophages, and enhanced both the intracellular expression of adaptor protein MyD88 and the inducible phosphorylation of IκBα and NF-κB. These findings provide additional information toward elucidating the mechanism by which HCMV potentiates bacteria-induced NF-κB-mediated macrophage inflammatory responses, thereby enhancing organ inflammation in HCMV-infected tissues.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Citomegalovirus/fisiología , Macrófagos/inmunología , Células Cultivadas , Citocinas/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/inmunología , Macrófagos/virología , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Transducción de Señal , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 5/inmunología , Replicación Viral
4.
Biochem J ; 467(3): 517-27, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25742174

RESUMEN

The apoA-I (apolipoprotein A-I) mimetic peptide 4F favours the differentiation of human monocytes to an alternatively activated M2 phenotype. The goal of the present study was to test whether the 4F-mediated differentiation of MDMs (monocyte-derived macrophages) requires the induction of an oxidative metabolic programme. 4F treatment induced several genes in MDMs that play an important role in lipid metabolism, including PPARγ (peroxisome-proliferator-activated receptor γ) and CD36. Addition of 4F was associated with a significant increase in FA (fatty acid) uptake and oxidation compared with vehicle treatment. Mitochondrial respiration was assessed by measurement of the OCR (oxygen-consumption rate). 4F increased basal and ATP-linked OCR as well as maximal uncoupled mitochondrial respiration. These changes were associated with a significant increase in ΔΨm (mitochondrial membrane potential). The increase in metabolic activity in 4F-treated MDMs was attenuated by etomoxir, an inhibitor of mitochondrial FA uptake. Finally, addition of the PPARγ antagonist T0070907 to 4F-treated MDMs reduced the expression of CD163 and CD36, cell-surface markers for M2 macrophages, and reduced basal and ATP-linked OCR. These results support our hypothesis that the 4F-mediated differentiation of MDMs to an anti-inflammatory phenotype is due, in part, to an increase in FA uptake and mitochondrial oxidative metabolism.


Asunto(s)
Apolipoproteína A-I/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Péptidos/farmacología , Antiinflamatorios/farmacología , Benzamidas/farmacología , Materiales Biomiméticos/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , Metabolismo Energético , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Macrófagos/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Consumo de Oxígeno , PPAR gamma/antagonistas & inhibidores , Piridinas/farmacología
5.
J Virol ; 88(13): 7683-8, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24741097

RESUMEN

We report that primary human vaginal dendritic cells (DCs) display a myeloid phenotype and express CD4, CCR5, and CXCR4. Vaginal CD13(+) CD11c(+) DCs rapidly and efficiently bound transmitted/founder (T/F) CCR5-tropic (R5) viruses, transported them through explanted vaginal mucosa, and transmitted them in trans to vaginal and blood lymphocytes. Vaginal myeloid DCs may play a key role in capturing and disseminating T/F R5 HIV-1 in vivo and are candidate "gatekeeper" cells in HIV-1 transmission.


Asunto(s)
Células Dendríticas/virología , Infecciones por VIH/transmisión , VIH-1/patogenicidad , Células Mieloides/virología , Linfocitos T/virología , Vagina/virología , Células Cultivadas , Células Dendríticas/metabolismo , Femenino , Infecciones por VIH/metabolismo , Humanos , Células Mieloides/metabolismo , Receptores CCR5/metabolismo , Linfocitos T/metabolismo , Vagina/metabolismo
6.
FASEB J ; 28(5): 2214-24, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24509909

RESUMEN

After Helicobacter pylori infection in humans, gastric epithelial cells (GECs) undergo apoptosis due to stimulation by the bacteria or inflammatory cytokines. In this study, we assessed the expression and function of brain angiogenesis inhibitor 1 (BAI1) in the engulfment of apoptotic GECs using human tissue and cells. After induction of apoptosis by H. pylori or camptothecin, there was a 5-fold increase in the binding of apoptotic GECs to THP-1 cells or peripheral blood monocyte-derived macrophages as assayed by confocal microscopy or conventional and imaging flow cytometry. Binding was impaired 95% by pretreating apoptotic cells with annexin V, underscoring the requirement for phosphatidylserine recognition. The phosphatidylserine receptor BAI1 was expressed in human gastric biopsy specimens and gastric phagocytes. To confirm the role of BAI1 in apoptotic cell clearance, the functional domain of BAI1 was used as a competitive inhibitor or BAI1 expression was inhibited by small interfering RNA. Both approaches decreased binding and engulfment >40%. Exposing THP-1 cells to apoptotic cells inhibited IL-6 production from 1340 to <364 pg/ml; however, this decrease was independent of phagocytosis. We conclude that recognition of apoptotic cells by BAI1 contributes to their clearance in the human gastric mucosa and this is associated with anti-inflammatory effects.


Asunto(s)
Proteínas Angiogénicas/metabolismo , Apoptosis , Células Epiteliales/metabolismo , Infecciones por Helicobacter/metabolismo , Fagocitos/metabolismo , Línea Celular , Técnicas de Cocultivo , Citocinas/metabolismo , Células Epiteliales/microbiología , Mucosa Gástrica/citología , Mucosa Gástrica/microbiología , Gastritis/metabolismo , Regulación de la Expresión Génica , Helicobacter pylori , Humanos , Inflamación , Macrófagos/citología , Macrófagos/metabolismo , Monocitos/citología , Fagocitos/citología , Fagocitosis , Receptores de Superficie Celular/química , Receptores Acoplados a Proteínas G , Estómago/citología , Estómago/microbiología
7.
J Immunol ; 190(12): 6626-34, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23686492

RESUMEN

Increased apoptotic death of gastric epithelial cells is a hallmark of Helicobacter pylori infection, and altered epithelial cell turnover is an important contributor to gastric carcinogenesis. To address the fate of apoptotic gastric epithelial cells and their role in H. pylori mucosal disease, we investigated phagocyte clearance of apoptotic gastric epithelial cells in H. pylori infection. Human gastric mononuclear phagocytes were analyzed for their ability to take up apoptotic epithelial cells (AECs) in vivo using immunofluorescence analysis. We then used primary human gastric epithelial cells induced to undergo apoptosis by exposure to live H. pylori to study apoptotic cell uptake by autologous monocyte-derived macrophages. We show that HLA-DR(+) mononuclear phagocytes in human gastric mucosa contain cytokeratin-positive and TUNEL-positive AEC material, indicating that gastric phagocytes are involved in AEC clearance. We further show that H. pylori both increased apoptosis in primary gastric epithelial cells and decreased phagocytosis of the AECs by autologous monocyte-derived macrophages. Reduced macrophage clearance of apoptotic cells was mediated in part by H. pylori-induced macrophage TNF-α, which was expressed at higher levels in H. pylori-infected, compared with uninfected, gastric mucosa. Importantly, we show that H. pylori-infected gastric mucosa contained significantly higher numbers of AECs and higher levels of nonphagocytosed TUNEL-positive apoptotic material, consistent with a defect in apoptotic cell clearance. Thus, as shown in other autoimmune and chronic inflammatory diseases, insufficient phagocyte clearance may contribute to the chronic and self-perpetuating inflammation in human H. pylori infection.


Asunto(s)
Apoptosis/fisiología , Células Epiteliales/patología , Infecciones por Helicobacter/inmunología , Leucocitos Mononucleares/inmunología , Macrófagos/inmunología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Mucosa Gástrica/citología , Mucosa Gástrica/inmunología , Infecciones por Helicobacter/patología , Helicobacter pylori , Humanos , Etiquetado Corte-Fin in Situ , Fagocitosis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Mucosal Immunol ; 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38945396

RESUMEN

Intestinal stromal cells (SCs), which synthesize the extracellular matrix that gives the mucosa its structure, are newly appreciated to play a role in mucosal inflammation. Here we show that human intestinal vimentin+CD90+SMA- SCs synthesize retinoic acid (RA) at levels equivalent to intestinal epithelial cells, a function in the human intestine previously attributed exclusively to epithelial cells. Crohn's disease SCs (Crohn's SCs), however, synthesized markedly less RA than SCs from healthy intestine (Normal SCs). We also show that microbe-stimulated Crohn's SCs, which are more inflammatory than stimulated Normal SCs, induced less RA-regulated differentiation of mucosal DCS (circulating pre-DCs and monocyte-derived DCs), leading to the generation of more potent inflammatory IFN-γhi/IL-17hi T cells than Normal SCs. Explaining these results, Crohn's SCs expressed more DHRS3, a retinaldehyde reductase that inhibits retinol conversion to retinal, and thus synthesized less RA than Normal SCs. These findings uncover a microbe-SC-DC crosstalk in which luminal microbes induce Crohn's disease SCs to initiate and perpetuate inflammation through impaired synthesis of RA.

9.
PLoS Pathog ; 7(5): e1002060, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21637819

RESUMEN

Tissue macrophages are derived exclusively from blood monocytes, which as monocyte-derived macrophages support HIV-1 replication. However, among human tissue macrophages only intestinal macrophages are non-permissive to HIV-1, suggesting that the unique microenvironment in human intestinal mucosa renders lamina propria macrophages non-permissive to HIV-1. We investigated this hypothesis using blood monocytes and intestinal extracellular matrix (stroma)-conditioned media (S-CM) to model the exposure of newly recruited monocytes and resident macrophages to lamina propria stroma, where the cells take up residence in the intestinal mucosa. Exposure of monocytes to S-CM blocked up-regulation of CD4 and CCR5 expression during monocyte differentiation into macrophages and inhibited productive HIV-1 infection in differentiated macrophages. Importantly, exposure of monocyte-derived macrophages simultaneously to S-CM and HIV-1 also inhibited viral replication, and sorted CD4+ intestinal macrophages, a proportion of which expressed CCR5+, did not support HIV-1 replication, indicating that the non-permissiveness to HIV-1 was not due to reduced receptor expression alone. Consistent with this conclusion, S-CM also potently inhibited replication of HIV-1 pseudotyped with vesicular stomatitis virus glycoprotein, which provides CD4/CCR5-independent entry. Neutralization of TGF-ß in S-CM and recombinant TGF-ß studies showed that stromal TGF-ß inhibited macrophage nuclear translocation of NF-κB and HIV-1 replication. Thus, the profound inability of intestinal macrophages to support productive HIV-1 infection is likely the consequence of microenvironmental down-regulation of macrophage HIV-1 receptor/coreceptor expression and NF-κB activation.


Asunto(s)
Antígenos CD4/metabolismo , Regulación hacia Abajo/fisiología , VIH-1/fisiología , Mucosa Intestinal/virología , Macrófagos/metabolismo , FN-kappa B/metabolismo , Receptores CCR5/metabolismo , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , VIH-1/aislamiento & purificación , VIH-1/patogenicidad , Humanos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/farmacología , Replicación Viral/efectos de los fármacos , Replicación Viral/fisiología
10.
Arterioscler Thromb Vasc Biol ; 32(11): 2631-9, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22982462

RESUMEN

OBJECTIVE: The apolipoprotein A-I (apoA-I) mimetic peptide 4F favors the differentiation of human monocytes to an anti-inflammatory phenotype and attenuates lipopolysaccharide (LPS)-induced inflammatory responses. We investigated the effects of LPS on the Toll-like receptor (TLR) signaling pathway in 4F-differentiated monocyte-derived macrophages. METHODS AND RESULTS: Monocyte-derived macrophages were pretreated with 4F or vehicle for 7 days. 4F downregulated cell-surface TLRs (4, 5, and 6) as determined by flow cytometry. 4F attenuated the LPS-dependent upregulation of genes encoding TLR1, 2, and 6 and genes of the MyD88-dependent (CD14, MyD88, TRAF6, interleukin-1 receptor-associated kinase 4, and inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase beta) and MyD88-independent (interferon regulatory factor 3, TANK-binding kinase 1, and Toll-interleukin 1 receptor domain-containing adaptor-inducing interferon-ß) pathways as determined by microarray analysis and quantitative reverse transcriptase polymerase chain reaction. Functional analyses of monocyte-derived macrophages showed that 4F reduced LPS-dependent TLR4 recycling, phosphorylation of nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha, activation and translocation of nuclear factor-κB and inhibited the secretion of tumor necrosis factor-α and interleukin-6 induced by LPS or lipoteichoic acid. These changes were associated with depletion of cellular cholesterol and caveolin, components of membrane lipid rafts. CONCLUSIONS: These data suggest that disruption of rafts by 4F alters the assembly of TLR-ligand complexes in cell membranes and inhibits proinflammatory gene expression in monocyte-derived macrophages, thus attenuating the responsiveness of macrophages to LPS.


Asunto(s)
Antiinflamatorios/farmacología , Apolipoproteína A-I/farmacología , Inflamación/prevención & control , Macrófagos/efectos de los fármacos , Péptidos/farmacología , Receptores Toll-Like/efectos de los fármacos , Transporte Activo de Núcleo Celular , Caveolina 1/metabolismo , Células Cultivadas , Colesterol/metabolismo , Citocinas/genética , Citocinas/metabolismo , Regulación hacia Abajo , Citometría de Flujo , Perfilación de la Expresión Génica/métodos , Humanos , Inflamación/genética , Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/inmunología , Microdominios de Membrana/efectos de los fármacos , Microdominios de Membrana/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Transcripción Genética/efectos de los fármacos
11.
JCI Insight ; 8(19)2023 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-37643018

RESUMEN

The role of aberrant glycosylation in pancreatic ductal adenocarcinoma (PDAC) remains an under-investigated area of research. In this study, we determined that ST6 ß-galactoside α2,6 sialyltransferase 1 (ST6GAL1), which adds α2,6-linked sialic acids to N-glycosylated proteins, was upregulated in patients with early-stage PDAC and was further increased in advanced disease. A tumor-promoting function for ST6GAL1 was elucidated using tumor xenograft experiments with human PDAC cells. Additionally, we developed a genetically engineered mouse (GEM) model with transgenic expression of ST6GAL1 in the pancreas and found that mice with dual expression of ST6GAL1 and oncogenic KRASG12D had greatly accelerated PDAC progression compared with mice expressing KRASG12D alone. As ST6GAL1 imparts progenitor-like characteristics, we interrogated ST6GAL1's role in acinar to ductal metaplasia (ADM), a process that fosters neoplasia by reprogramming acinar cells into ductal, progenitor-like cells. We verified ST6GAL1 promotes ADM using multiple models including the 266-6 cell line, GEM-derived organoids and tissues, and an in vivo model of inflammation-induced ADM. EGFR is a key driver of ADM and is known to be activated by ST6GAL1-mediated sialylation. Importantly, EGFR activation was dramatically increased in acinar cells and organoids from mice with transgenic ST6GAL1 expression. These collective results highlight a glycosylation-dependent mechanism involved in early stages of pancreatic neoplasia.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Ratones , Animales , Neoplasias Pancreáticas/patología , Páncreas/patología , Carcinoma Ductal Pancreático/patología , Receptores ErbB/genética , Metaplasia/patología , Sialiltransferasas/genética , beta-D-Galactósido alfa 2-6-Sialiltransferasa , Antígenos CD
12.
Gastroenterology ; 141(3): 929-38, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21699795

RESUMEN

BACKGROUND & AIMS: Mucosal dendritic cells (DCs) play a key role in initiating the T-helper (Th)1 response to Helicobacter pylori. To further elucidate the mucosal response to H pylori, we examined whether gastric stromal factors condition DCs to support tolerance to H pylori, analogous to intestinal stromal factor-driven macrophage tolerance to commensal bacteria. METHODS: To model mucosal DC development, we isolated and cultured cell-depleted human stroma/extracellular matrix from fresh gastric and intestinal mucosa to generate stroma-conditioned media. We then analyzed the capacity of stroma-conditioned media-treated monocyte-derived DCs and primary human gastric and intestinal DCs pulsed in vitro with H pylori to induce T-cell proliferation and interferon gamma secretion. RESULTS: Stromal factors in gastric mucosa suppressed H pylori-stimulated DC activation and the ability of DCs to drive a Th1 proliferative and cytokine response to H pylori. The ability of gastric stromal factors to down-regulate DC function was similar to that of intestinal stromal factors and was independent of transforming growth factor ß, prostaglandin E2, interleukin (IL)-10, and thymic stromal lymphopoietin. Stroma-conditioned media-induced reduction in DC-stimulated Th1 responses was associated with reduced DC release of IL-12. CONCLUSIONS: Gastric stromal factors down-regulate DC responsiveness to H pylori, resulting in a dampened gastric Th1 response. We speculate that stroma-induced down-regulation of DC function contributes to the permissiveness of both gastric and intestinal mucosa to colonization by persistent residential microbes.


Asunto(s)
Comunicación Celular/fisiología , Células Dendríticas/citología , Helicobacter pylori/fisiología , Intestino Delgado/citología , Estómago/citología , Células del Estroma/citología , Células TH1/citología , Proliferación Celular , Células Cultivadas , Quimiocinas CXC/metabolismo , Quimiocinas CXC/farmacología , Medios de Cultivo Condicionados/farmacología , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Dinoprostona/metabolismo , Mucosa Gástrica/metabolismo , Humanos , Interleucina-10/metabolismo , Intestino Delgado/efectos de los fármacos , Intestino Delgado/metabolismo , Estómago/efectos de los fármacos , Células del Estroma/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Linfopoyetina del Estroma Tímico
13.
J Immunol ; 184(5): 2289-96, 2010 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-20139273

RESUMEN

We previously identified that autoreactive B cells from BXD2 mice can be targeted by IL-17, leading to upregulation of the expression of regulators of G-protein signaling (Rgs) genes that facilitated the development of spontaneous germinal centers. Little is known about the signaling pathway used by IL-17 to upregulate RGS. In the current study, we found that IL-17 rapidly activates the canonical NF-kappaB signaling pathway and that BXD2 B cells exhibit higher basal and activated phosphorylated p65 levels than B6 or BXD2-Il17ra(-/-) B cells. Inhibition of p65 phosphorylation downregulated RGS16 expression and abrogated the IL-17-induced chemotactic arrest of B cells in response to CXCL12. Knockdown of TNFR-associated factor 6 or NF-kappaB activator 1 in 70Z/3 pre-B cells led to decreased Rgs16 expression, indicating that both of these two genes are involved in IL-17-mediated activation of NF-kappaB signaling in B cells. These findings identify the signaling pathway regulated by IL-17 to contribute to the development of spontaneous germinal centers in autoimmune BXD2 mice.


Asunto(s)
Linfocitos B/metabolismo , Interleucina-17/farmacología , FN-kappa B/metabolismo , Proteínas RGS/metabolismo , Transducción de Señal/efectos de los fármacos , Transporte Activo de Núcleo Celular/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Autoinmunidad , Linfocitos B/citología , Linfocitos B/inmunología , Western Blotting , Línea Celular , Núcleo Celular/metabolismo , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Proteínas RGS/genética , Interferencia de ARN , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo , Regulación hacia Arriba/efectos de los fármacos
14.
J Immunol ; 184(7): 3648-55, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20208001

RESUMEN

Monostratified epithelial cells translocate HIV type 1 (HIV-1) from the apical to the basolateral surface via vesicular transcytosis. Because acutely transmitted HIV-1 is almost exclusively CCR5-tropic and human intestinal epithelial cells preferentially transcytose CCR5-tropic virus, we established epithelial monolayers using polarized HT-29 cells transduced to express CCR5, and an explant system using normal human rectal mucosa, to characterize biological parameters of epithelial cell transcytosis of HIV-1 and assess antiviral Ab blockade of transcytosis. The amount of cell-free HIV-1 transcytosed through the epithelial monolayer increased linearly in relation to the amount of virus applied to the apical surface, indicating transcytosis efficiency was constant (r(2) = 0.9846; p < 0.0001). The efficiency of HIV-1 transcytosis ranged between 0.05 and 1.21%, depending on the virus strain, producer cell type and gp120 V1-V3 loop signature. Inoculation of HIV-1 neutralizing Abs to the immunodominant region (7B2) or the conserved membrane proximal external region (2F5) of gp41 or to cardiolipin (IS4) onto the apical surface of epithelial monolayers prior to inoculation of virus significantly reduced HIV-1 transcytosis. 2F5 was the most potent of these IgG1 Abs. Dimeric IgA and monomeric IgA, but not polymeric IgM, 2F5 Abs also blocked HIV-1 transcytosis across the epithelium and, importantly, across explanted normal human rectal mucosa, with monomeric IgA substantially more potent than dimeric IgA in effecting transcytosis blockade. These findings underscore the potential role of transcytosis blockade in the prevention of HIV-1 transmission across columnar epithelium such as that of the rectum.


Asunto(s)
Fármacos Anti-VIH/inmunología , Anticuerpos Antivirales/inmunología , Proteína gp41 de Envoltorio del VIH/inmunología , Infecciones por VIH/transmisión , VIH-1/inmunología , Mucosa Intestinal/virología , Fármacos Anti-VIH/farmacología , Anticuerpos Antivirales/farmacología , Células Epiteliales/inmunología , Células Epiteliales/virología , Infecciones por VIH/prevención & control , Células HT29 , Humanos , Mucosa Intestinal/inmunología , Receptores CCR5/inmunología , Recto/inmunología , Recto/virología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
J Biol Chem ; 285(25): 19593-604, 2010 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-20388715

RESUMEN

Human intestinal macrophages contribute to tissue homeostasis in noninflamed mucosa through profound down-regulation of pro-inflammatory cytokine release. Here, we show that this down-regulation extends to Toll-like receptor (TLR)-induced cytokine release, as intestinal macrophages expressed TLR3-TLR9 but did not release cytokines in response to TLR-specific ligands. Likely contributing to this unique functional profile, intestinal macrophages expressed markedly down-regulated adapter proteins MyD88 and Toll interleukin receptor 1 domain-containing adapter-inducing interferon beta, which together mediate all TLR MyD88-dependent and -independent NF-kappaB signaling, did not phosphorylate NF-kappaB p65 or Smad-induced IkappaBalpha, and did not translocate NF-kappaB into the nucleus. Importantly, transforming growth factor-beta released from intestinal extracellular matrix (stroma) induced identical down-regulation in the NF-kappaB signaling and function of blood monocytes, the exclusive source of intestinal macrophages. Our findings implicate stromal transforming growth factor-beta-induced dysregulation of NF-kappaB proteins and Smad signaling in the differentiation of pro-inflammatory blood monocytes into noninflammatory intestinal macrophages.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Proteínas I-kappa B/metabolismo , Inflamación , Mucosa Intestinal/metabolismo , Macrófagos/metabolismo , FN-kappa B/metabolismo , Proteínas Smad/metabolismo , Citocinas/metabolismo , Inhibidores Enzimáticos/química , Humanos , Monocitos/metabolismo , Inhibidor NF-kappaB alfa , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
16.
Am J Physiol Cell Physiol ; 298(6): C1538-48, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20219948

RESUMEN

HDL and its major protein component apolipoprotein A-I (apoA-I) exert anti-inflammatory effects, inhibit monocyte chemotaxis/adhesion, and reduce vascular macrophage content in inflammatory conditions. In this study, we tested the hypothesis that the apoA-I mimetic 4F modulates the function of monocyte-derived macrophages (MDMs) by regulating the expression of key cell surface receptors on MDMs. Primary human monocytes and THP-1 cells were treated with 4F, apoA-I, or vehicle for 7 days and analyzed for expression of cell surface markers, adhesion to human endothelial cells, phagocytic function, cholesterol efflux capacity, and lipid raft organization. 4F and apoA-I treatment decreased the expression of HLA-DR, CD86, CD11b, CD11c, CD14, and Toll-like receptor-4 (TLR-4) compared with control cells, suggesting the induction of monocyte differentiation. Both treatments abolished LPS-induced mRNA for monocyte chemotactic protein-1 (MCP-1), macrophage inflammatory protein-1 (MIP-1), regulated on activation, normal T-expressed and presumably secreted (RANTES), IL-6, and TNF-alpha but significantly upregulated LPS-induced IL-10 expression. Moreover, 4F and apoA-I induced a 90% reduction in the expression of CD49d, a ligand for the VCAM-1 receptor, with a concurrent decrease in monocyte adhesion (55% reduction) to human endothelial cells and transendothelial migration (34 and 27% for 4F and apoA-I treatments) compared with vehicle treatment. In addition, phagocytosis of dextran-FITC beads was inhibited by 4F and apoA-I, a response associated with reduced expression of CD32. Finally, 4F and apoA-I stimulated cholesterol efflux from MDMs, leading to cholesterol depletion and disruption of lipid rafts. These data provide evidence that 4F, similar to apoA-I, induces profound functional changes in MDMs, possibly due to differentiation to an anti-inflammatory phenotype.


Asunto(s)
Antiinflamatorios/farmacología , Apolipoproteína A-I/farmacología , Macrófagos/efectos de los fármacos , Antígenos CD/metabolismo , Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Colesterol/metabolismo , Citocinas/genética , Citocinas/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Antígenos HLA-DR/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Rodamiento de Leucocito/efectos de los fármacos , Lipopolisacáridos/farmacología , Macrófagos/inmunología , Macrófagos/metabolismo , Microdominios de Membrana/efectos de los fármacos , Microdominios de Membrana/metabolismo , Imitación Molecular , Fagocitosis/efectos de los fármacos , Fenotipo , ARN Mensajero/metabolismo
17.
Kidney Int ; 78(6): 550-60, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20555320

RESUMEN

Monocyte and macrophage markers are among the most highly overexpressed genes in cpk mouse kidneys with severely progressive renal cystic disease. We show here that one of these markers, CD14, is abnormally transcribed, activated and shed in cystic kidneys. However, these abnormalities were not associated with an increased number of interstitial CD14-positive mononuclear cells. Instead, we found that most non-cystic and cystic renal tubular epithelia were CD14-positive; even distal nephron-derived principal cells. Cd14 was significantly overexpressed in the kidneys of 5-day-old cpk mice and further increased as the disease progressed. In the cpk model with variable rates of cystic kidney enlargement (due to an intercross of two distinct genetic backgrounds), Cd14 expression positively correlated with kidney volume, exceeding the correlation with MCP-1, an established marker of autosomal-dominant polycystic kidney disease (ADPKD). In 16 patients with ADPKD, the baseline urinary CD14 level showed some tendency to correlate with the 2-year change in total kidney volume; however, the tendency was not statistically significant. But the association was significant when the analysis was confined to males. Clearly more studies need to be done to evaluate the utility of CD14 as a marker for outcomes in ADPKD.


Asunto(s)
Enfermedades Renales Quísticas/patología , Receptores de Lipopolisacáridos/análisis , Riñón Poliquístico Autosómico Dominante/patología , Índice de Severidad de la Enfermedad , Animales , Animales Recién Nacidos , Progresión de la Enfermedad , Femenino , Humanos , Riñón/química , Riñón/metabolismo , Enfermedades Renales Quísticas/diagnóstico , Receptores de Lipopolisacáridos/metabolismo , Masculino , Ratones , Tamaño de los Órganos , Riñón Poliquístico Autosómico Dominante/diagnóstico , Factores Sexuales , Distribución Tisular
18.
J Virol ; 83(7): 3258-67, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19153236

RESUMEN

Mucosal surfaces play a major role in human immunodeficiency virus type 1 (HIV-1) transmission and pathogenesis, and yet the role of lamina propria macrophages in mucosal HIV-1 infection has received little investigative attention. We report here that vaginal and intestinal macrophages display distinct phenotype and HIV-1 permissiveness profiles. Vaginal macrophages expressed the innate response receptors CD14, CD89, CD16, CD32, and CD64 and the HIV-1 receptor/coreceptors CD4, CCR5, and CXCR4, similar to monocytes. Consistent with this phenotype, green fluorescent protein-tagged R5 HIV-1 entered macrophages in explanted vaginal mucosa as early as 30 min after inoculation of virus onto the epithelium, and purified vaginal macrophages supported substantial levels of HIV-1 replication by a panel of highly macrophage-tropic R5 viruses. In sharp contrast, intestinal macrophages expressed no detectable, or very low levels of, innate response receptors and HIV-1 receptor/coreceptors and did not support HIV-1 replication, although virus occasionally entered macrophages in intestinal tissue explants. Thus, vaginal, but not intestinal, macrophages are monocyte-like and permissive to R5 HIV-1 after the virus has translocated across the epithelium. These findings suggest that genital and gut macrophages have different roles in mucosal HIV-1 pathogenesis and that vaginal macrophages play a previously underappreciated but potentially important role in mucosal HIV-1 infection in the female genital tract.


Asunto(s)
VIH-1/crecimiento & desarrollo , Mucosa Intestinal/virología , Macrófagos/virología , Vagina/virología , Antígenos CD/análisis , Femenino , Humanos , Macrófagos/química , Técnicas de Cultivo de Órganos , Receptores del VIH/análisis
19.
Gastroenterology ; 134(2): 491-9, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18242215

RESUMEN

BACKGROUND & AIMS: Helicobacter pylori infection in children infrequently causes gastroduodenal mucosal ulceration. Because H pylori induces T-cell dependent gastric inflammation in adults and T regulatory (Treg) cells suppress T-cell-dependent pathology, we evaluated gastric histopathology and Treg cell responses in H pylori-infected children and adults. METHODS: Gastric tissue from 36 children and 79 adults with abdominal symptoms in Santiago, Chile, was evaluated prospectively for H pylori bacteria and histopathology using the Sydney classification and Treg responses using immunoassay, immunohistochemistry, and real-time polymerase chain reaction. RESULTS: Eighteen (50%) of the children and 51 (65%) of the adults were infected with H pylori. Children and adults were colonized with similar levels of H pylori. However, the level of gastritis in the children was reduced substantially compared with that of the adults (P < .05). Coincident with reduced gastric inflammation, the number of Treg cells and levels of Treg cytokines (transforming growth factor [TGF]-beta1 and interleukin-10) were increased markedly in the gastric mucosa of H pylori-infected children compared with that of infected adults (P < .03 and < .05, respectively). Also, H pylori infection in the children was associated with markedly increased levels of gastric TGF-beta1 and interleukin-10 messenger RNA. Importantly, gastric TGF-beta1 in H pylori-infected children localized predominantly to mucosal CD25(+) and Foxp3(+) cells, indicating a Treg source for the TGF-beta1. CONCLUSIONS: Gastric pathology is reduced and local Treg cell responses are increased in H pylori-infected children compared with infected adults, suggesting that gastric Treg cell responses down-regulate the inflammation and ulceration induced by H pylori in children.


Asunto(s)
Gastritis/inmunología , Gastritis/virología , Infecciones por Helicobacter/inmunología , Helicobacter pylori/patogenicidad , Linfocitos T Reguladores/patología , Linfocitos T Reguladores/fisiología , Adolescente , Adulto , Envejecimiento/patología , Proliferación Celular , Niño , Chile , Regulación hacia Abajo , Femenino , Factores de Transcripción Forkhead/metabolismo , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patología , Mucosa Gástrica/virología , Gastritis/etiología , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/patología , Humanos , Interleucina-10/metabolismo , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Masculino , Estudios Prospectivos , ARN Mensajero/metabolismo , Linfocitos T Reguladores/virología , Factor de Crecimiento Transformador beta1/metabolismo
20.
Sci Rep ; 9(1): 14724, 2019 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-31604984

RESUMEN

The intestinal microbiota is critical for maintaining homeostasis. Dysbiosis, an imbalance in the microbial community, contributes to the susceptibility of several diseases. Many factors are known to influence gut microbial composition, including diet. We have previously shown that fecal immunoglobulin (Ig) A levels are decreased in mice fed a diet free of aryl hydrocarbon receptor (AhR) ligands. Here, we hypothesize this IgA decrease is secondary to diet-induced dysbiosis. We assigned mice to a conventional diet, an AhR ligand-free diet, or an AhR ligand-free diet supplemented with the dietary AhR ligand indole-3-carbinol (I3C). We observed a global alteration of fecal microbiota upon dietary AhR ligand deprivation. Compared to mice on the conventional diet, family Erysipelotrichaceae was enriched in the feces of mice on the AhR ligand-free diet but returned to normal levels upon dietary supplementation with I3C. Faecalibaculum rodentium, an Erysipelotrichaceae species, depleted its growth media of AhR ligands. Cultured fecal bacteria from mice on the AhR ligand-free diet, but not the other two diets, were able to alter IgA levels in vitro, as was F. rodentium alone. Our data point to the critical role of AhR dietary ligands in shaping the composition and proper functioning of gut microbiota.


Asunto(s)
Dieta/métodos , Suplementos Dietéticos , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/inmunología , Receptores de Hidrocarburo de Aril , Animales , ADN Ribosómico/genética , Dieta/efectos adversos , Disbiosis/etiología , Heces/química , Heces/microbiología , Firmicutes/genética , Células HCT116 , Humanos , Inmunoglobulina A/análisis , Indoles , Ligandos , Ratones , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA