Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
1.
J Virol ; 98(6): e0071224, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38780246

RESUMEN

Within the first 15 minutes of infection, herpes simplex virus 1 immediate early proteins repurpose cellular RNA polymerase (Pol II) for viral transcription. An important role of the viral-infected cell protein 27 (ICP27) is to facilitate viral pre-mRNA processing and export viral mRNA to the cytoplasm. Here, we use precision nuclear run-on followed by deep sequencing (PRO-seq) to characterize transcription of a viral ICP27 null mutant. At 1.5 and 3 hours post infection (hpi), we observed increased total levels of Pol II on the mutant viral genome and accumulation of Pol II downstream of poly A sites indicating increased levels of initiation and processivity. By 6 hpi, Pol II accumulation on specific mutant viral genes was higher than that on wild-type virus either at or upstream of poly A signals, depending on the gene. The PRO-seq profile of the ICP27 mutant on late genes at 6 hpi was similar but not identical to that caused by treatment with flavopiridol, a known inhibitor of RNA processivity. This pattern was different from PRO-seq profiles of other α gene mutants and upon inhibition of viral DNA replication with PAA. Together, these results indicate that ICP27 contributes to the repression of aberrant viral transcription at 1.5 and 3 hpi by inhibiting initiation and decreasing RNA processivity. However, ICP27 is needed to enhance processivity on most late genes by 6 hpi in a mechanism distinguishable from its role in viral DNA replication.IMPORTANCEWe developed and validated the use of a processivity index for precision nuclear run-on followed by deep sequencing data. The processivity index calculations confirm infected cell protein 27 (ICP27) induces downstream of transcription termination on certain host genes. The processivity indices and whole gene probe data implicate ICP27 in transient immediate early gene-mediated repression, a process that also requires ICP4, ICP22, and ICP0. The data indicate that ICP27 directly or indirectly regulates RNA polymerase (Pol II) initiation and processivity on specific genes at specific times post infection. These observations support specific and varied roles for ICP27 in regulating Pol II activity on viral genes in addition to its known roles in post transcriptional mRNA processing and export.


Asunto(s)
Genoma Viral , Herpesvirus Humano 1 , Proteínas Inmediatas-Precoces , Mutación , ARN Polimerasa II , Transcripción Viral , Animales , Humanos , Línea Celular , Chlorocebus aethiops , Regulación Viral de la Expresión Génica/efectos de los fármacos , Genes Virales/genética , Genoma Viral/genética , Herpes Simple/virología , Herpes Simple/genética , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiología , Proteínas Inmediatas-Precoces/deficiencia , Proteínas Inmediatas-Precoces/genética , Poli A/genética , Poli A/metabolismo , ARN Polimerasa II/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Células Vero , Transcripción Viral/efectos de los fármacos , Transcripción Viral/genética , Replicación Viral/genética
2.
J Virol ; 97(10): e0096023, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37754762

RESUMEN

IMPORTANCE: Infection with herpes simplex virus 1 (HSV-1) leads to lifelong infection due to the virus's remarkable ability to control transcription of its own genome, resulting in two transcriptional programs: lytic (highly active) and latent (restricted). The lytic program requires immediate early (IE) proteins to first repress transcription of late viral genes, which then undergo sequential de-repression, leading to a specific sequence of gene expression. Here, we show that the IE ICP4 functions to regulate the cascade by limiting RNA polymerase initiation at immediate early times. However, late viral genes that initiate too early in the absence of ICP4 do not yield mRNA as transcription stalls within gene bodies. It follows that other regulatory steps intercede to prevent elongation of genes at the incorrect time, demonstrating the precise control HSV-1 exerts over its own transcription.


Asunto(s)
Regulación Viral de la Expresión Génica , Herpesvirus Humano 1 , Proteínas Inmediatas-Precoces , Transcripción Genética , Humanos , Genes Virales/genética , Herpes Simple/virología , Herpesvirus Humano 1/genética , Proteínas Inmediatas-Precoces/deficiencia , Proteínas Inmediatas-Precoces/metabolismo , Iniciación de la Transcripción Genética , Elongación de la Transcripción Genética , Terminación de la Transcripción Genética
3.
Cell Rep ; 37(1): 109785, 2021 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-34610303

RESUMEN

A hallmark of type 2 diabetes (T2D) is hepatic resistance to insulin's glucose-lowering effects. The serum- and glucocorticoid-regulated family of protein kinases (SGK) is activated downstream of mechanistic target of rapamycin complex 2 (mTORC2) in response to insulin in parallel to AKT. Surprisingly, despite an identical substrate recognition motif to AKT, which drives insulin sensitivity, pathological accumulation of SGK1 drives insulin resistance. Liver-specific Sgk1-knockout (Sgk1Lko) mice display improved glucose tolerance and insulin sensitivity and are protected from hepatic steatosis when fed a high-fat diet. Sgk1 promotes insulin resistance by inactivating AMP-activated protein kinase (AMPK) via phosphorylation on inhibitory site AMPKαSer485/491. We demonstrate that SGK1 is dominant among SGK family kinases in regulation of insulin sensitivity, as Sgk1, Sgk2, and Sgk3 triple-knockout mice have similar increases in hepatic insulin sensitivity. In aggregate, these data suggest that targeting hepatic SGK1 may have therapeutic potential in T2D.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Hígado/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Animales , Diabetes Mellitus Tipo 2/patología , Dieta Alta en Grasa , Proteína Forkhead Box O1/metabolismo , Glucosa/metabolismo , Proteínas Inmediatas-Precoces/deficiencia , Proteínas Inmediatas-Precoces/genética , Insulina/metabolismo , Resistencia a la Insulina , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Guía de Kinetoplastida/metabolismo , Transducción de Señal
4.
J Neuroinflammation ; 18(1): 86, 2021 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-33812385

RESUMEN

BACKGROUND: Subcortical ischemic vascular dementia, one of the major subtypes of vascular dementia, is characterized by lacunar infarcts and white matter lesions caused by chronic cerebral hypoperfusion. In this study, we used a mouse model of bilateral common carotid artery stenosis (BCAS) to investigate the role of B-cell translocation gene 2 (BTG2), an antiproliferation gene, in the white matter glial response to chronic cerebral hypoperfusion. METHODS: Btg2-/- mice and littermate wild-type control mice underwent BCAS or sham operation. Behavior phenotypes were assessed by open-field test and Morris water maze test. Brain tissues were analyzed for the degree of white matter lesions and glial changes. To further confirm the effects of Btg2 deletion on proliferation of glial cells in vitro, BrdU incorporation was investigated in mixed glial cells derived from wild-type and Btg2-/- mice. RESULTS: Relative to wild-type mice with or without BCAS, BCAS-treated Btg2-/- mice exhibited elevated spontaneous locomotor activity and poorer spatial learning ability. Although the severities of white matter lesions did not significantly differ between wild-type and Btg2-/- mice after BCAS, the immunoreactivities of GFAP, a marker of astrocytes, and Mac2, a marker of activated microglia and macrophages, in the white matter of the optic tract were higher in BCAS-treated Btg2-/- mice than in BCAS-treated wild-type mice. The expression level of Gfap was also significantly elevated in BCAS-treated Btg2-/- mice. In vitro analysis showed that BrdU incorporation in mixed glial cells in response to inflammatory stimulation associated with cerebral hypoperfusion was higher in Btg2-/- mice than in wild-type mice. CONCLUSION: BTG2 negatively regulates glial cell proliferation in response to cerebral hypoperfusion, resulting in behavioral changes.


Asunto(s)
Circulación Cerebrovascular/genética , Eliminación de Gen , Proteínas Inmediatas-Precoces/deficiencia , Proteínas Inmediatas-Precoces/genética , Neuroglía/metabolismo , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Sustancia Blanca/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuroglía/patología , Sustancia Blanca/patología
5.
Anal Cell Pathol (Amst) ; 2019: 3013765, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31815093

RESUMEN

Inflammation plays a pivotal role in the development of pulmonary arterial hypertension (PAH). Meanwhile, serum glucocorticoid-regulated kinase-1 (SGK1) has been considered to be an important factor in the regulation of inflammation in some vascular disease. However, the role of SGK1 in hypoxia-induced inflammation and PAH is still unknown. WT and SGK1-/- mice were exposed to chronic hypoxia to induce PAH. The quantitative PCR and immunohistochemistry were used to determine the expression of SGK1. The right ventricular hypertrophy index (RVHI), RV/BW ratio, right ventricle systolic pressure (RVSP), and percentage of muscularised vessels and medical wall thickness were measured to evaluate PAH development. The infiltration of macrophages and localization of SGK1 on cells were examined by histological analysis. The effects of SGK1 on macrophage function and cytokine expression were assessed by comparing WT and SGK1-/- macrophages in vitro. SGK1 has high expression in hypoxia-induced PAH. Deficiency of SGK1 prevented the development of hypoxia-induced PAH and inhibited macrophage infiltration in the lung. In addition, SGK1 knockout inhibited the expression of proinflammatory cytokines in macrophages. SGK1-induced macrophage activation and proinflammatory response contributes to the development of PAH in hypoxia-treated mice. Thus, SGK1 might be considered a promising target for PAH treatment.


Asunto(s)
Hipertensión Pulmonar/complicaciones , Hipertensión Pulmonar/enzimología , Hipoxia/complicaciones , Hipoxia/enzimología , Proteínas Inmediatas-Precoces/metabolismo , Activación de Macrófagos , Macrófagos/enzimología , Macrófagos/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Médula Ósea/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Hipertensión Pulmonar/fisiopatología , Hipoxia/fisiopatología , Proteínas Inmediatas-Precoces/deficiencia , Inflamación/patología , Pulmón/enzimología , Pulmón/patología , Pulmón/fisiopatología , Masculino , Ratones , Proteínas Serina-Treonina Quinasas/deficiencia , Remodelación Vascular
6.
Invest Ophthalmol Vis Sci ; 60(10): 3398-3406, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31387116

RESUMEN

Purpose: We previously have reported that ICP22, an immediate early gene of herpes simplex virus type 1 (HSV-1), binds to the CD80 promoter to suppress CD80 expression in antigen-presenting cells, leading to reduced T-cell function and protection. In contrast, overexpression of CD80 exacerbates corneal scarring (CS) in ocularly infected mice. In this study we tested the hypothesis that the absence of ICP22 could increase disease severity. Methods: To test our hypothesis, BALB/c mice were ocularly infected after corneal scarification with a recombinant HSV-1 lacking the ICP22 gene with its parental wild-type (WT) virus (KOS) as a control. Virus replication in the eye, CS, angiogenesis, latency, and reactivation between ICP22 null virus and WT KOS were determined. In addition, expression of IL-2, IL-4, IFN-γ, IFN-α, granzyme A, granzyme B, and perforin by CD4 and CD8 T cells in corneas of infected mice on days 3, 5, 7, 10, 14, 21, and 28 postinfection were determined by flow cytometry. Results: We found similar levels of eye disease and angiogenesis in mice following corneal scarification and ocular infection with the ICP22 null virus or parental WT virus despite reduced virus replication in the eye and reduced latency and reactivation in mice ocularly infected with ICP22 null virus. The similar level of eye disease in ICP22 null virus- and WT virus-infected mice correlated with expression of various proinflammatory cytokines that infiltrated the eye after HSV-1 infection. Conclusions: Our study identified a critical role for ICP22 in HSV-1 pathogenicity and suggests that HSV-1-associated CS is more dependent on host immune responses to infection than to virus replication in the eye. Thus, HSV-1 as means of survival uses ICP22 as a mechanism of immune escape that protects the host from increased pathology.


Asunto(s)
Sustancia Propia/virología , Herpesvirus Humano 1/fisiología , Proteínas Inmediatas-Precoces/fisiología , Queratitis Herpética/inmunología , Latencia del Virus/fisiología , Animales , Antígeno B7-1/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Movimiento Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Proteínas Inmediatas-Precoces/deficiencia , Queratitis Herpética/patología , Ratones , Ratones Endogámicos BALB C , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Lágrimas/virología , Replicación Viral/fisiología
7.
PLoS Pathog ; 14(8): e1007264, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30138450

RESUMEN

Herpes Simplex Virus 1 (HSV1) is amongst the most clinically advanced oncolytic virus platforms. However, efficient and sustained viral replication within tumours is limiting. Rapamycin can stimulate HSV1 replication in cancer cells, but active-site dual mTORC1 and mTORC2 (mammalian target of rapamycin complex 1 and 2) inhibitors (asTORi) were shown to suppress the virus in normal cells. Surprisingly, using the infected cell protein 0 (ICP0)-deleted HSV1 (HSV1-dICP0), we found that asTORi markedly augment infection in cancer cells and a mouse mammary cancer xenograft. Mechanistically, asTORi repressed mRNA translation in normal cells, resulting in defective antiviral response but also inhibition of HSV1-dICP0 replication. asTORi also reduced antiviral response in cancer cells, however in contrast to normal cells, transformed cells and cells transduced to elevate the expression of eukaryotic initiation factor 4E (eIF4E) or to silence the repressors eIF4E binding proteins (4E-BPs), selectively maintained HSV1-dICP0 protein synthesis during asTORi treatment, ultimately supporting increased viral replication. Our data show that altered eIF4E/4E-BPs expression can act to promote HSV1-dICP0 infection under prolonged mTOR inhibition. Thus, pharmacoviral combination of asTORi and HSV1 can target cancer cells displaying dysregulated eIF4E/4E-BPs axis.


Asunto(s)
Herpes Simple/patología , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 1/genética , Proteínas Inmediatas-Precoces/genética , Neoplasias/virología , Inhibidores de Proteínas Quinasas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Dominio Catalítico/efectos de los fármacos , Proteínas de Ciclo Celular , Células Cultivadas , Chlorocebus aethiops , Factor 4E Eucariótico de Iniciación/genética , Factor 4E Eucariótico de Iniciación/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Herpes Simple/complicaciones , Herpes Simple/genética , Humanos , Proteínas Inmediatas-Precoces/deficiencia , Ratones , Neoplasias/complicaciones , Neoplasias/genética , Neoplasias/patología , Organismos Modificados Genéticamente , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/química , Ubiquitina-Proteína Ligasas/deficiencia , Células Vero
8.
J Clin Invest ; 128(7): 3024-3040, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29889103

RESUMEN

Medial vascular calcification, associated with enhanced mortality in chronic kidney disease (CKD), is fostered by osteo-/chondrogenic transdifferentiation of vascular smooth muscle cells (VSMCs). Here, we describe that serum- and glucocorticoid-inducible kinase 1 (SGK1) was upregulated in VSMCs under calcifying conditions. In primary human aortic VSMCs, overexpression of constitutively active SGK1S422D, but not inactive SGK1K127N, upregulated osteo-/chondrogenic marker expression and activity, effects pointing to increased osteo-/chondrogenic transdifferentiation. SGK1S422D induced nuclear translocation and increased transcriptional activity of NF-κB. Silencing or pharmacological inhibition of IKK abrogated the osteoinductive effects of SGK1S422D. Genetic deficiency, silencing, and pharmacological inhibition of SGK1 dissipated phosphate-induced calcification and osteo-/chondrogenic transdifferentiation of VSMCs. Aortic calcification, stiffness, and osteo-/chondrogenic transdifferentiation in mice following cholecalciferol overload were strongly reduced by genetic knockout or pharmacological inhibition of Sgk1 by EMD638683. Similarly, Sgk1 deficiency blunted vascular calcification in apolipoprotein E-deficient mice after subtotal nephrectomy. Treatment of human aortic smooth muscle cells with serum from uremic patients induced osteo-/chondrogenic transdifferentiation, effects ameliorated by EMD638683. These observations identified SGK1 as a key regulator of vascular calcification. SGK1 promoted vascular calcification, at least partly, via NF-κB activation. Inhibition of SGK1 may, thus, reduce the burden of vascular calcification in CKD.


Asunto(s)
Proteínas Inmediatas-Precoces/metabolismo , Miocitos del Músculo Liso/metabolismo , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Calcificación Vascular/metabolismo , Animales , Benzamidas/farmacología , Transdiferenciación Celular/efectos de los fármacos , Transdiferenciación Celular/genética , Transdiferenciación Celular/fisiología , Células Cultivadas , Condrogénesis/efectos de los fármacos , Condrogénesis/genética , Condrogénesis/fisiología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Hidrazinas/farmacología , Proteínas Inmediatas-Precoces/deficiencia , Proteínas Inmediatas-Precoces/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Noqueados para ApoE , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Osteogénesis/fisiología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Insuficiencia Renal/metabolismo , Insuficiencia Renal/patología , Transducción de Señal , Calcificación Vascular/etiología , Calcificación Vascular/patología
9.
Viruses ; 9(8)2017 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-28783105

RESUMEN

Infection with herpes simplex virus-1 (HSV-1) brings numerous changes in cellular gene expression. Levels of most host mRNAs are reduced, limiting synthesis of host proteins, especially those involved in antiviral defenses. The impact of HSV-1 on host microRNAs (miRNAs), an extensive network of short non-coding RNAs that regulate mRNA stability/translation, remains largely unexplored. Here we show that transcription of the miR-183 cluster (miR-183, miR-96, and miR-182) is selectively induced by HSV-1 during productive infection of primary fibroblasts and neurons. ICP0, a viral E3 ubiquitin ligase expressed as an immediate-early protein, is both necessary and sufficient for this induction. Nuclear exclusion of ICP0 or removal of the RING (really interesting new gene) finger domain that is required for E3 ligase activity prevents induction. ICP0 promotes the degradation of numerous host proteins and for the most part, the downstream consequences are unknown. Induction of the miR-183 cluster can be mimicked by depletion of host transcriptional repressors zinc finger E-box binding homeobox 1 (ZEB1)/-crystallin enhancer binding factor 1 (δEF1) and zinc finger E-box binding homeobox 2 (ZEB2)/Smad-interacting protein 1 (SIP1), which we establish as new substrates for ICP0-mediated degradation. Thus, HSV-1 selectively stimulates expression of the miR-183 cluster by ICP0-mediated degradation of ZEB transcriptional repressors.


Asunto(s)
Herpesvirus Humano 1/enzimología , Interacciones Huésped-Patógeno , MicroARNs/genética , Ubiquitina-Proteína Ligasas/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Núcleo Celular , Células Cultivadas , Fibroblastos/virología , Regulación de la Expresión Génica , Herpesvirus Humano 1/genética , Humanos , Proteínas Inmediatas-Precoces/deficiencia , Proteínas Inmediatas-Precoces/genética , Proteínas del Tejido Nervioso/genética , Neuronas/virología , Unión Proteica , Proteolisis , Proteínas de Unión al ARN/genética , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/genética , Replicación Viral , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética
11.
Cell Physiol Biochem ; 39(4): 1295-306, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27606670

RESUMEN

BACKGROUND: Serum & glucocorticoid inducible kinase (SGK1) regulates several ion channels, including amiloride sensitive epithelial Na+ channel (ENaC). SGK1 and ENaC in the luminal endometrium epithelium, are critically involved in embryo implantation, although little is known about their regulation. The present study explored whether SGK1 and ENaC are modulated by LEFTYA, a negative regulator of uterine receptivity. METHODS: Expression levels were determined by qRT-PCR and Western blotting, ENaC channel activity by whole cell patch clamp and transepithelial current by Ussing chamber experiments. RESULTS: Treatment of Ishikawa cells, an endometrial adenocarcinoma model cell line of endometrial epithelial cells, with LEFTYA rapidly up-regulated SGK1 and ENaC transcript and protein levels. Induction of ENaC in response to LEFTYA was blunted upon co-treatment with the SGK1 inhibitor EMD638683. ENaC levels also significantly upregulated upon expression of a constitutively active, but not a kinase dead, SGK1 mutant in Ishikawa cells. LEFTYA increased amiloride sensitive Na+-currents in Ishikawa cells and amiloride sensitive transepithelial current across the murine endometrium. Furthermore, LEFTYA induced the expression of ENaC in the endometrium of wild-type but not of Sgk1-deficient mice. CONCLUSIONS: LEFTYA regulates the expression and activity of ENaC in endometrial epithelial cells via SGK1. Aberrant regulation of SGK1 and ENaC by LEFTYA could contribute to the pathogenesis of unexplained infertility.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Canales Epiteliales de Sodio/genética , Proteínas Inmediatas-Precoces/genética , Factores de Determinación Derecha-Izquierda/farmacología , Proteínas Serina-Treonina Quinasas/genética , ARN Mensajero/genética , Amilorida/farmacología , Animales , Benzamidas/farmacología , Línea Celular Tumoral , Cámaras de Difusión de Cultivos , Endometrio/citología , Endometrio/efectos de los fármacos , Endometrio/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Canales Epiteliales de Sodio/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Hidrazinas/farmacología , Proteínas Inmediatas-Precoces/antagonistas & inhibidores , Proteínas Inmediatas-Precoces/deficiencia , Factores de Determinación Derecha-Izquierda/genética , Factores de Determinación Derecha-Izquierda/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Placa-Clamp , Cultivo Primario de Células , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/deficiencia , ARN Mensajero/metabolismo , Transducción de Señal
12.
Sci Rep ; 6: 26786, 2016 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-27226390

RESUMEN

The use of engineered viral strains such as gene therapy vectors and oncolytic viruses (OV) to selectively destroy cancer cells is poised to make a major impact in the clinic and revolutionize cancer therapy. In particular, several studies have shown that OV therapy is safe and well tolerated in humans and can infect a broad range of cancers. Yet in clinical studies OV therapy has highly variable response rates. The heterogeneous nature of tumors is widely accepted to be a major obstacle for OV therapeutics and highlights a need for strategies to improve viral replication efficacy. Here, we describe the development of a new class of small molecules for selectively enhancing OV replication in cancer tissue. Medicinal chemistry studies led to the identification of compounds that enhance multiple OVs and gene therapy vectors. Lead compounds increase OV growth up to 2000-fold in vitro and demonstrate remarkable selectivity for cancer cells over normal tissue ex vivo and in vivo. These small molecules also demonstrate enhanced stability with reduced electrophilicity and are highly tolerated in animals. This pharmacoviral approach expands the scope of OVs to include resistant tumors, further potentiating this transformative therapy. It is easily foreseeable that this approach can be applied to therapeutically enhance other attenuated viral vectors.


Asunto(s)
Furanos/farmacología , Herpesvirus Humano 1/efectos de los fármacos , Viroterapia Oncolítica/métodos , Virus Oncolíticos/efectos de los fármacos , Virus de la Estomatitis Vesicular Indiana/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Adenocarcinoma/terapia , Animales , Línea Celular Tumoral , Neoplasias del Colon/terapia , Evaluación Preclínica de Medicamentos , Estabilidad de Medicamentos , Femenino , Glutatión/análisis , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiología , Proteínas Inmediatas-Precoces/deficiencia , Proteínas Inmediatas-Precoces/genética , Ratones , Ratones Endogámicos BALB C , Virus Oncolíticos/genética , Virus Oncolíticos/fisiología , Suero , Estimulación Química , Relación Estructura-Actividad , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/genética , Virus de la Estomatitis Vesicular Indiana/genética , Virus de la Estomatitis Vesicular Indiana/fisiología , Proteínas de la Matriz Viral/deficiencia , Proteínas de la Matriz Viral/genética
13.
Sci Rep ; 6: 24135, 2016 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-27063893

RESUMEN

Chronic inflammation plays a crucial role in the pathogenesis of obesity and insulin resistance. However, the primary mediators that affect energy homeostasis remain ill defined. Here, we report an unexpected role for immediate early response gene X-1 (IEX-1), a downstream target of NF-κB, in energy metabolism. We found that IEX-1 expression was highly induced in white adipose tissue (WAT) in both epidydmal and subcutaneous depots but not in interscapular brown adipose tissue (BAT) in mice fed a high fat diet (HFD). Null mutation of IEX-1 protected mice against HFD-induced adipose and hepatic inflammation, hepatic steatosis, and insulin resistance. Unexpectedly, IEX-1 knockout (IEX-1(-/-)) mice gained markedly less weight on HFD for 20 weeks as compared to wild-type (WT) littermates (37 ± 3 versus 48 ± 2 gm) due to increased energy expenditure. Mechanistically, we showed that IEX-1 deficiency induced browning and activated thermogenic genes program in WAT but not in BAT by promoting alternative activation of adipose macrophages. Consequently, IEX-1(-/-) mice exhibited enhanced thermogenesis (24 ± 0.1 versus 22 ± 0.1 kcal/hour/kg in WT mice) explaining increased energy expenditure and lean phenotype in these mice. In conclusion, the present study suggests that IEX-1 is a novel physiological regulator of energy homeostasis via its action in WAT.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Dieta Alta en Grasa , Proteínas Inmediatas-Precoces/genética , Obesidad/etiología , Animales , Calorimetría , Metabolismo Energético , Hígado Graso/etiología , Expresión Génica , Prueba de Tolerancia a la Glucosa , Proteínas Inmediatas-Precoces/deficiencia , Inmunohistoquímica , Inmunofenotipificación , Insulina/sangre , Resistencia a la Insulina , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , ARN Mensajero/metabolismo , Termogénesis
14.
Am J Physiol Renal Physiol ; 311(2): F330-42, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27009335

RESUMEN

The stimulation of postprandial K(+) clearance involves aldosterone-independent and -dependent mechanisms. In this context, serum- and glucocorticoid-induced kinase (SGK)1, a ubiquitously expressed kinase, is one of the primary aldosterone-induced proteins in the aldosterone-sensitive distal nephron. Germline inactivation of SGK1 suggests that this kinase is fundamental for K(+) excretion under conditions of K(+) load, but the specific role of renal SGK1 remains elusive. To avoid compensatory mechanisms that may occur during nephrogenesis, we used inducible, nephron-specific Sgk1(Pax8/LC1) mice to assess the role of renal tubular SGK1 in K(+) regulation. Under a standard diet, these animals exhibited normal K(+) handling. When challenged by a high-K(+) diet, they developed severe hyperkalemia accompanied by a defect in K(+) excretion. Molecular analysis revealed reduced neural precursor cell expressed developmentally downregulated protein (NEDD)4-2 phosphorylation and total expression. γ-Epithelial Na(+) channel (ENaC) expression and α/γENaC proteolytic processing were also decreased in mutant mice. Moreover, with no lysine kinase (WNK)1, which displayed in control mice punctuate staining in the distal convoluted tubule and diffuse distribution in the connecting tubule/cortical colleting duct, was diffused in the distal convoluted tubule and less expressed in the connecting tubule/collecting duct of Sgk(Pax8/LC1) mice. Moreover, Ste20-related proline/alanine-rich kinase phosphorylation, and Na(+)-Cl(-) cotransporter phosphorylation/apical localization were reduced in mutant mice. Consistent with the altered WNK1 expression, increased renal outer medullary K(+) channel apical localization was observed. In conclusion, our data suggest that renal tubular SGK1 is important in the regulation of K(+) excretion via the control of NEDD4-2, WNK1, and ENaC.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Canales Epiteliales de Sodio/metabolismo , Proteínas Inmediatas-Precoces/deficiencia , Proteínas Inmediatas-Precoces/genética , Antígenos de Histocompatibilidad Menor/metabolismo , Potasio/orina , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Bloqueadores/farmacología , Dieta , Regulación de la Expresión Génica , Túbulos Renales/metabolismo , Masculino , Ratones , Ratones Noqueados , Ubiquitina-Proteína Ligasas Nedd4 , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , Canales de Potasio de Rectificación Interna/inmunología , Potasio en la Dieta/farmacología , Proteína Quinasa Deficiente en Lisina WNK 1
15.
J Bone Miner Res ; 31(3): 573-84, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26391411

RESUMEN

Bone homeostasis is maintained by the synergistic actions of bone-resorbing osteoclasts and bone-forming osteoblasts. Although interferon-related developmental regulator 1 (Ifrd1) has been identified as a transcriptional coactivator/repressor in various cells, little attention has been paid to its role in osteoblastogenesis and bone homeostasis thus far. Here, we show that Ifrd1 is a critical mediator of both the cell-autonomous regulation of osteoblastogenesis and osteoblast-dependent regulation of osteoclastogenesis. Osteoblast-specific deletion of murine Ifrd1 increased bone formation and decreased bone resorption, causing high bone mass. Ifrd1 deficiency enhanced osteoblast differentiation and maturation along with increased expression of Runx2 and osterix (Osx). Mechanistically, Ifrd1 deficiency increased the acetylation status of p65, a component of NF-κB, at residues K122 and K123 via the attenuation of the interaction between p65 and histone deacetylase (HDAC). This led to the nuclear export of p65 and a decrease in NF-κB-dependent Smad7 expression and the subsequent enhancement of Smad1/Smad5/Smad8-dependent transcription. Moreover, a high bone mass phenotype in the osteoblast-specific deletion of Ifrd1 was markedly rescued by the introduction of one Osx-floxed allele but not of Runx2-floxed allele. Coculture experiments revealed that Ifrd1-deficient osteoblasts have a higher osteoprotegerin (OPG) expression and a lower ability to support osteoclastogenesis. Ifrd1 deficiency attenuated the interaction between ß-catenin and HDAC, subsequently increasing the acetylation of ß-catenin at K49, leading to its nuclear accumulation and the activation of the ß-catenin-dependent transcription of OPG. Collectively, the expression of Ifrd1 in osteoblasts repressed osteoblastogenesis and activated osteoclastogenesis through modulating the NF-κB/Smad/Osx and ß-catenin/OPG pathways, respectively. These findings suggest that Ifrd1 has a pivotal role in bone homeostasis through its expression in osteoblasts in vivo and represents a therapeutic target for bone diseases.


Asunto(s)
Resorción Ósea/metabolismo , Resorción Ósea/patología , Proteínas Inmediatas-Precoces/metabolismo , Proteínas de la Membrana/metabolismo , Osteoblastos/metabolismo , Osteogénesis , Transcripción Genética , Acetilación , Animales , Huesos/patología , Diferenciación Celular , Núcleo Celular/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Eliminación de Gen , Histona Desacetilasas/metabolismo , Homeostasis , Proteínas Inmediatas-Precoces/deficiencia , Proteínas de la Membrana/deficiencia , Ratones Noqueados , FN-kappa B/metabolismo , Tamaño de los Órganos , Osteoblastos/patología , Osteoprotegerina/metabolismo , Fenotipo , Proteínas Smad/metabolismo , Factor de Transcripción Sp7 , Factores de Transcripción/metabolismo , beta Catenina/metabolismo
16.
J Thromb Haemost ; 13(7): 1325-34, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25944668

RESUMEN

BACKGROUND: Platelet secretion is critical to development of acute thrombotic occlusion. Platelet dense granules contain a variety of important hemostatically active substances. Nevertheless, biogenesis of platelet granules is poorly understood. OBJECTIVES: Serum- and glucocorticoid-inducible kinase 1 (SGK1) has been shown to be highly expressed in platelets and megakaryocytes, but its role in the regulation of platelet granule biogenesis and its impact on thrombosis has not been investigated so far. METHODS AND RESULTS: Electron microscopy analysis of the platelet ultrastructure revealed a significant reduction in the number and packing of dense granules in platelets lacking SGK1 (sgk1(-/-) ). In sgk1(-/-) platelets serotonin content was significantly reduced and activation-dependent secretion of ATP, serotonin and CD63 significantly impaired. In vivo adhesion after carotis ligation was significantly decreased in platelets lacking SGK1 and occlusive thrombus formation after FeCl3 -induced vascular injury was significantly diminished in sgk1(-/-) mice. Transcript levels and protein abundance of dense granule biogenesis regulating GTPase Rab27b were significantly reduced in sgk1(-/-) platelets without affecting Rab27b mRNA stability. In MEG-01 cells transfection with constitutively active (S422) (D) SGK1 but not with inactive (K127) (N) SGK1 significantly enhanced Rab27b mRNA levels. Sgk1(-/-) megakaryocytes show significantly reduced expression of Rab27b and serotonin/CD63 levels compared with sgk1(+/+) megakaryocytes. Proteome analysis identified nine further vesicular transport proteins regulated by SGK1, which may have an impact on impaired platelet granule biogenesis in sgk1(-/-) platelets independent of Rab27b. CONCLUSIONS: The present observations identify SGK1 as a novel powerful regulator of platelet dense granule biogenesis, platelet secretion and thrombus formation. SGK1 is at least partially effective because it regulates transcription of Rab27b in megakaryocytes.


Asunto(s)
Plaquetas/enzimología , Traumatismos de las Arterias Carótidas/enzimología , Gránulos Citoplasmáticos/enzimología , Proteínas Inmediatas-Precoces/sangre , Activación Plaquetaria , Proteínas Serina-Treonina Quinasas/sangre , Vesículas Secretoras/enzimología , Trombosis/enzimología , Adenosina Trifosfato/sangre , Adenosina Trifosfato/metabolismo , Animales , Plaquetas/metabolismo , Plaquetas/ultraestructura , Traumatismos de las Arterias Carótidas/sangre , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/patología , Células Cultivadas , Gránulos Citoplasmáticos/metabolismo , Gránulos Citoplasmáticos/ultraestructura , Modelos Animales de Enfermedad , Femenino , Genotipo , Proteínas Inmediatas-Precoces/deficiencia , Proteínas Inmediatas-Precoces/genética , Masculino , Megacariocitos/enzimología , Megacariocitos/metabolismo , Ratones Noqueados , Fenotipo , Agregación Plaquetaria , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , ARN Mensajero/metabolismo , Vesículas Secretoras/metabolismo , Vesículas Secretoras/ultraestructura , Serotonina/sangre , Serotonina/metabolismo , Transducción de Señal , Tetraspanina 30/sangre , Tetraspanina 30/metabolismo , Trombosis/sangre , Trombosis/genética , Trombosis/patología , Factores de Tiempo , Transfección , Regulación hacia Arriba , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo
17.
Arterioscler Thromb Vasc Biol ; 35(3): 547-57, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25614279

RESUMEN

OBJECTIVE: Atherosclerosis, an inflammatory disease of arterial vessel walls, requires migration and matrix metalloproteinase (MMP)-9-dependent invasion of monocytes/macrophages into the vascular wall. MMP-9 expression is stimulated by transcription factor nuclear factor-κB, which is regulated by inhibitor κB (IκB) and thus IκB kinase. Regulators of nuclear factor-κB include serum- and glucocorticoid-inducible kinase 1 (SGK1). The present study explored involvement of SGK1 in vascular inflammation and atherogenesis. APPROACH AND RESULTS: Gene-targeted apolipoprotein E (ApoE)-deficient mice without (apoe(-/-)sgk1(+/+)) or with (apoe(-/-)sgk1(-/-)) additional SGK1 knockout received 16-week cholesterol-rich diet. According to immunohistochemistry atherosclerotic lesions in aorta and carotid artery, vascular CD45(+) leukocyte infiltration, Mac-3(+) macrophage infiltration, vascular smooth muscle cell content, MMP-2, and MMP-9 positive areas in atherosclerotic tissue were significantly less in apoe(-/-)sgk1(-/-)mice than in apoe(-/-)sgk1(+/+)mice. As determined by Boyden chamber, thioglycollate-induced peritonitis and air pouch model, migration of SGK1-deficient CD11b(+)F4/80(+) macrophages was significantly diminished in vitro and in vivo. Zymographic MMP-2 and MMP-9 production, MMP-9 activity and invasion through matrigel in vitro were significantly less in sgk1(-/-) than in sgk1(+/+)macrophages and in control plasmid-transfected or inactive (K127N)SGK1-transfected than in constitutively active (S422D)SGK1-transfected THP-1 cells. Confocal microscopy revealed reduced macrophage number and macrophage MMP-9 content in plaques of apoe(-/-)sgk1(-/-) mice. In THP-1 cells, MMP-inhibitor GM6001 (25 µmol/L) abrogated (S422D)SGK1-induced MMP-9 production and invasion. According to reverse transcription polymerase chain reaction, MMP-9 transcript levels were significantly reduced in sgk1(-/-)macrophages and strongly upregulated in (S422D)SGK1-transfected THP-1 cells compared with control plasmid-transfected or (K127N)SGK1-transfected THP-1 cells. According to immunoblotting and confocal microscopy, phosphorylation of IκB kinase and inhibitor κB and nuclear translocation of p50 were significantly lower in sgk1(-/-)macrophages than in sgk1(+/+)macrophages and significantly higher in (S422D)SGK1-transfected THP-1 cells than in control plasmid-transfected or (K127N)SGK1-transfected THP-1 cells. Treatment of (S422D)SGK1-transfected THP-1 cells with IκB kinase-inhibitor BMS-345541 (10 µmol/L) abolished (S422D)SGK1-induced increase of MMP-9 transcription and gelatinase activity. CONCLUSIONS: SGK1 plays a pivotal role in vascular inflammation during atherogenesis. SGK1 participates in the regulation of monocyte/macrophage migration and MMP-9 transcription via regulation of nuclear factor-κB.


Asunto(s)
Enfermedades de la Aorta/enzimología , Aterosclerosis/enzimología , Enfermedades de las Arterias Carótidas/enzimología , Quimiotaxis , Proteínas Inmediatas-Precoces/metabolismo , Inflamación/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte Activo de Núcleo Celular , Animales , Aorta/enzimología , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/patología , Arterias Carótidas/enzimología , Arterias Carótidas/patología , Enfermedades de las Arterias Carótidas/genética , Enfermedades de las Arterias Carótidas/patología , Línea Celular , Modelos Animales de Enfermedad , Regulación Enzimológica de la Expresión Génica , Humanos , Quinasa I-kappa B/metabolismo , Proteínas I-kappa B/metabolismo , Proteínas Inmediatas-Precoces/deficiencia , Proteínas Inmediatas-Precoces/genética , Inflamación/genética , Inflamación/patología , Macrófagos/enzimología , Macrófagos/patología , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Subunidad p50 de NF-kappa B/metabolismo , Peritonitis/inducido químicamente , Peritonitis/enzimología , Peritonitis/genética , Placa Aterosclerótica , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal , Tioglicolatos , Transcripción Genética , Transfección , Remodelación Vascular
18.
Pflugers Arch ; 467(9): 1965-74, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25394886

RESUMEN

Duchenne muscular dystrophy (DMD) is a human genetic disease characterized by fibrosis and severe muscle weakness. Currently, there is no effective treatment available to prevent progressive fibrosis in skeletal muscles. The serum- and glucocorticoid-inducible kinase SGK1 regulates a variety of physiological functions and participates in fibrosis stimulation. Here, we investigated whether SGK1 influences structure, function and/or fibrosis of the muscles from the mdx mouse, an animal model for DMD. As expected, mdx muscles showed the typical pathological features of muscular dystrophy including fiber size variations, central nuclei of muscle fibers, fibrosis in the diaphragm, and force reduction by 30-50 %. Muscles from sgk1 (-/-) mice were histologically overall intact and specific force was only slightly reduced compared to wild-type muscles. Surprisingly, soleus and diaphragm muscles of mdx/sgk1 (-/-) mice displayed forces close to wild-type levels. Most muscle fibers of the double mutants contained central nuclei, but fibrosis was not observed in any of the tested limb and diaphragm muscles. We conclude that the sole lack of SGK1 in mouse muscle does not lead to pronounced changes in muscle structure and function. However, dystrophin-deficient mdx muscle seems to benefit from SGK1 deficiency. SGK1 appears to be an important enzyme in the process of fibrotic remodeling and subsequent weakness of dystrophin-deficient mouse muscle.


Asunto(s)
Proteínas Inmediatas-Precoces/metabolismo , Fuerza Muscular/fisiología , Músculo Esquelético/fisiología , Distrofia Muscular de Duchenne/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Modelos Animales de Enfermedad , Fibrosis/metabolismo , Proteínas Inmediatas-Precoces/deficiencia , Masculino , Ratones , Ratones Endogámicos mdx , Ratones Noqueados , Músculo Esquelético/patología , Proteínas Serina-Treonina Quinasas/deficiencia
19.
Int Immunopharmacol ; 23(2): 658-63, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25466275

RESUMEN

Sepsis, a leading cause of mortality in intensive care units worldwide, is often a result of overactive and systemic inflammation following serious infections. We found that mice lacking immediate early responsive gene X-1 (IEX-1) were prone to lipopolysaccharide (LPS) -induced endotoxemia. A nonlethal dose of LPS provoked numerous aberrations in IEX-1 knockout (KO) mice including pancytopenia, increased serum aspartate aminotransferase (AST), and lung neutrophilia, concurrent with liver and kidney damage, followed by death. Given these results, in conjunction with a proven role for IEX-1 in the regulation of reactive oxygen species (ROS) homeostasis during stress, we pre-treated IEX-1 KO mice with Mitoquinone (MitoQ), a mitochondrion-based antioxidant prior to LPS injection. The treatment significantly reduced ROS formation in circulatory cells and protected against pancytopenia and multiple organ failure, drastically increasing the survival rate of IEX-1 KO mice challenged by this low dose of LPS. This study confirms significant contribution of mitochondrial ROS to the etiology of sepsis.


Asunto(s)
Antioxidantes/uso terapéutico , Endotoxemia/prevención & control , Endotoxinas/toxicidad , Proteínas Inmediatas-Precoces/deficiencia , Mitocondrias/efectos de los fármacos , Compuestos Organofosforados/uso terapéutico , Ubiquinona/análogos & derivados , Animales , Antioxidantes/administración & dosificación , Recuento de Células Sanguíneas , Interpretación Estadística de Datos , Endotoxemia/sangre , Endotoxemia/inducido químicamente , Endotoxemia/genética , Proteínas Inmediatas-Precoces/genética , Estimación de Kaplan-Meier , Riñón/efectos de los fármacos , Riñón/patología , Hígado/efectos de los fármacos , Hígado/patología , Pulmón/efectos de los fármacos , Pulmón/patología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones Noqueados , Mitocondrias/metabolismo , Compuestos Organofosforados/administración & dosificación , Especies Reactivas de Oxígeno/metabolismo , Ubiquinona/administración & dosificación , Ubiquinona/uso terapéutico
20.
Am J Physiol Gastrointest Liver Physiol ; 307(6): G642-54, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25059825

RESUMEN

Effective therapies are limited for patients with parenteral nutrition-dependent short bowel syndrome. We previously showed that intestinal expression of the transcriptional coregulator tetradecanoyl phorbol acetate-induced sequence 7 (tis7) is markedly increased during the adaptive response following massive small bowel resection and tis7 plays a role in normal gut lipid metabolism. Here, we further explore the functional implications of tis7 deletion in intestinal lipid metabolism and the adaptive response following small bowel resection. Intestinal tis7 transgenic (tis7(tg)), tis7(-/-), and wild-type (WT) littermates were subjected to 50% small bowel resection. Mice were fed a control or a high-saturated-fat (42% energy) diet for 21 days. Survival, body weight recovery, lipid absorption, mucosal lipid analysis, and the morphometric adaptive response were analyzed. Quantitative real-time PCR was performed to identify tis7 downstream gene targets. Postresection survival was markedly reduced in high-fat, but not control, diet-fed tis7(-/-) mice. Decreased survival was associated with anastomotic inflammation and intestinal obstruction postresection. High-fat, but not control, diet-fed tis7(-/-) mice had increased intestinal IL-6 expression. Intestinal lipid trafficking was altered in tis7(-/-) compared with WT mice postresection. In contrast, high-fat diet-fed tis7(tg) mice had improved survival postresection compared with WT littermates. High-fat diet feeding in the setting of tis7 deletion resulted in postresection anastomotic inflammation and small bowel obstruction. Tolerance of a calorie-rich, high-fat diet postresection may require tis7 and its target genes. The presence of luminal fat in the setting of tis7 deletion promotes an intestinal inflammatory response postresection.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Enteritis/etiología , Proteínas Inmediatas-Precoces/deficiencia , Obstrucción Intestinal/etiología , Intestino Delgado/metabolismo , Proteínas de la Membrana/deficiencia , Síndrome del Intestino Corto/complicaciones , Anastomosis Quirúrgica , Animales , Modelos Animales de Enfermedad , Enteritis/genética , Enteritis/metabolismo , Regulación de la Expresión Génica , Proteínas Inmediatas-Precoces/genética , Interleucina-6/metabolismo , Absorción Intestinal , Obstrucción Intestinal/genética , Obstrucción Intestinal/metabolismo , Intestino Delgado/cirugía , Metabolismo de los Lípidos , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Síndrome del Intestino Corto/genética , Síndrome del Intestino Corto/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...