Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
CEN Case Rep ; 10(4): 510-514, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33837952

RESUMEN

Glomerulopathy with Fibronectin Deposits (GFND) is a rare, autosomal dominant disease characterized by proteinuria, hematuria and progressive renal failure associated with glomerular deposition of fibronectin, frequently resulting in end-stage renal disease (ESRD). There is no established treatment for this condition beyond conservative measures such as blood pressure control and the use of angiotensin-converting enzyme (ACE) inhibitors. We present a case of GFND associated with progressive chronic kidney disease (CKD) and nephrotic range proteinuria showing a sustained response to prednisone treatment. A 27-year-old G2P2 Caucasian female presented with 3 g/day of proteinuria, serum creatinine (Cr) 0.7 mg/dL, inactive urinary sediment and normotension without medication. She was part of a large family with glomerular disease, including three members who died of cerebral hemorrhage or stroke in their thirties. The patient's kidney biopsy showed mesangial deposition of fibronectin consistent with GFND. No interstitial fibrosis was seen. Genotyping revealed the Y973C FN1 gene mutation. Despite maximal tolerable ACE inhibition, proteinuria increased to 4-6 g/g Cr and serum Cr increased to 1.0 mg/dL. She was treated with prednisone 60 mg (~ 1 mg/Kg) daily for 2 mos and then tapered by ~ 0.2 mg/Kg every month for 6 mos of total therapy. Proteinuria decreased to ~ 1 g/g Cr for > 5 years and serum Cr stabilized in the 1.2 mg/dL range with treatment. No significant side effects were encountered. In conclusion, this protocol should be considered in GFND patients with nephrotic range proteinuria despite maximal angiotensin system inhibition who have relatively preserved renal function.


Asunto(s)
Glomerulonefritis Membranoproliferativa/tratamiento farmacológico , Glucocorticoides/uso terapéutico , Prednisona/uso terapéutico , Adulto , Femenino , Glomerulonefritis Membranoproliferativa/genética , Glomerulonefritis Membranoproliferativa/patología , Humanos , Riñón/ultraestructura , Inducción de Remisión
2.
Sci Rep ; 9(1): 10019, 2019 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-31273219

RESUMEN

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

3.
PLoS One ; 13(11): e0206407, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30388136

RESUMEN

The aryl hydrocarbon receptor (AHR) is a ligand activated bHLH transcription factor that belongs to the Per-Arnt-Sim (PAS) superfamily of proteins involved in mediating responses to cellular environment regulating normal physiological and developmental pathways. The AHR binds a broad range of naturally derived and synthetic compounds, and plays a major role in mediating effects of certain environmental chemicals. Although our understanding of the physiological roles of the AHR in the immune system is evolving, there is little known about its role in hematopoiesis and hematopoietic diseases. Prior studies demonstrated that AHR null (AHR-KO) mice have impaired hematopoietic stem cell (HSC) function; they develop myeloproliferative changes in peripheral blood cells, and alterations in hematopoietic stem and progenitor cell populations in the bone marrow. We hypothesized mice lacking AHR expression only within hematopoietic cells (AHRVav1 mice) would develop similar changes. However, we did not observe a complete phenocopy of AHR-KO and AHRVav1 animals at 2 or 18 months of age. To illuminate the signaling mechanisms underlying the alterations in hematopoiesis observed in these mice, we sorted a population of cells highly enriched for HSC function (LSK cells: CD34-CD48-CD150+) and performed microarray analyses. Ingenuity Pathway and Gene Set Enrichment Analyses revealed that that loss of AHR within HSCs alters several gene and signaling networks important for HSC function. Differences in gene expression networks among HSCs from AHR-KO and AHRVav1 mice suggest that AHR in bone marrow stromal cells also contributes to HSC function. In addition, numerous studies have suggested a role for AHR in both regulation of hematopoietic cells, and in the development of blood diseases. More work is needed to define what these signals are, and how they act upon HSCs.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Eliminación de Gen , Células Madre Hematopoyéticas/metabolismo , Receptores de Hidrocarburo de Aril/deficiencia , Receptores de Hidrocarburo de Aril/genética , Transcriptoma/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células Madre Hematopoyéticas/citología , Ratones , Fenotipo , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal/genética
4.
Sci Rep ; 8(1): 1630, 2018 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-29374238

RESUMEN

Candida albicans is a diploid fungus and a predominant opportunistic human pathogen. Notably, C. albicans employs reversible chromosomal aneuploidies as a means of survival in adverse environments. We previously characterized transcription on the monosomic chromosome 5 (Ch5) that arises with adaptation to growth on the toxic sugar sorbose in the mutant Sor125(55). We now extend this analysis to the trisomic hybrid Ch4/7 within Sor125(55) and a diverse group of three mutants harboring a single Ch5. We find a similar pattern of transcriptional changes on either type of aneuploid chromosome within these mutants wherein expression of many genes follows chromosome ploidy, consistent with a direct mechanism to regulate genes important for adaptation to growth. In contrast, a significant number of genes are expressed at the disomic level, implying distinct mechanisms compensating for gene dose on monosomic or trisomic chromosomes consistent with maintaining cell homeostasis. Finally, we find evidence for an additional mechanism that elevates expression of genes on normal disomic Ch4 and Ch7 in mutants to levels commensurate with that found on the trisomic Ch4/7b in Sor125(55). Several of these genes are similarly differentially regulated among mutants, suggesting they play key functions in either maintaining aneuploidy or adaptation to growth conditions.


Asunto(s)
Adaptación Biológica , Aneuploidia , Candida albicans/genética , Cromosomas Fúngicos , Regulación de la Expresión Génica , Sorbosa/toxicidad , Transcripción Genética , Candida albicans/efectos de los fármacos
5.
Epigenetics Chromatin ; 10(1): 52, 2017 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-29115969

RESUMEN

After the publication of this work [1], it was noticed that an initial was missing from the author name: Jeffrey Hayes. His name should be written as: Jeffrey J. Hayes.

6.
Epigenetics Chromatin ; 10(1): 49, 2017 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-29061172

RESUMEN

BACKGROUND: The major human fungal pathogen Candida albicans possesses a diploid genome, but responds to growth in challenging environments by employing chromosome aneuploidy as an adaptation mechanism. For example, we have shown that C. albicans adapts to growth on the toxic sugar L-sorbose by transitioning to a state in which one chromosome (chromosome 5, Ch5) becomes monosomic. Moreover, analysis showed that while expression of many genes on the monosomic Ch5 is altered in accordance with the chromosome ploidy, expression of a large fraction of genes is increased to the normal diploid level, presumably compensating for gene dose. RESULTS: In order to understand the mechanism of the apparent dosage compensation, we now report genome-wide ChIP-microarray assays for a sorbose-resistant strain harboring a monosomic Ch5. These data show a significant chromosome-wide elevation in histone H4 acetylation on the mCh5, but not on any other chromosome. Importantly, strains lacking subunits of the NuA4 H4 histone acetyltransferase complex, orthologous to a complex previously shown in Drosophila to be associated with a similar gene dosage compensation mechanism, did not show an increase in H4 acetylation. Moreover, loss of NuA4 subunits severely compromised the adaptation to growth on sorbose. CONCLUSIONS: Our results are consistent with a model wherein chromosome-wide elevation of H4 acetylation mediated by the NuA4 complex plays a role in increasing gene expression in compensation for gene dose and adaption to growth in a toxic environment.


Asunto(s)
Farmacorresistencia Fúngica , Proteínas Fúngicas/metabolismo , Histona Acetiltransferasas/metabolismo , Histonas/metabolismo , Procesamiento Proteico-Postraduccional , Acetilación , Candida albicans/enzimología , Candida albicans/genética , Candida albicans/metabolismo , Compensación de Dosificación (Genética) , Proteínas Fúngicas/genética , Regulación Fúngica de la Expresión Génica , Histona Acetiltransferasas/genética , Monosomía
7.
PLoS One ; 11(5): e0155347, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27228053

RESUMEN

We have previously shown that regulatory T cells (Tregs) infiltrating follicular lymphoma lymph nodes are quantitatively and qualitatively different than those infiltrating normal and reactive nodes. To gain insight into how such Treg populations differ, we performed RNA sequence (RNAseq) analyses on flow sorted Tregs from all three sources. We identify several molecules that could contribute to the observed increased suppressive capacity of follicular lymphoma nodal tregs, including upregulation of CTLA-4, IL-10, and GITR, all confirmed by protein expression. In addition, we identify, and confirm functionally, a novel mechanism by which Tregs target to and accumulate within a human tumor microenvironment, through the down regulation of S1PR1, SELL (L-selectin) and CCR7, potentially resulting in greater lymph node retention. In addition we identify and confirm functionally the upregulation of the chemokine receptor CXCR5 as well as the secretion of the chemokines CXCL13 and IL-16 demonstrating the unique ability of the follicular derived Tregs to localize and accumulate within not only the malignant lymph node, but also localize and accumulate within the malignant B cell follicle itself. Such findings offer significant new insights into how follicular lymphoma nodal Tregs may contribute to the biology of follicular lymphoma and identify several novel therapeutic targets.


Asunto(s)
Movimiento Celular/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Ganglios Linfáticos/inmunología , Linfoma Folicular/inmunología , Proteínas de Neoplasias/inmunología , Linfocitos T Reguladores/inmunología , Transcriptoma/inmunología , Linfocitos B/inmunología , Linfocitos B/patología , Femenino , Perfilación de la Expresión Génica , Humanos , Ganglios Linfáticos/patología , Linfoma Folicular/patología , Masculino , Linfocitos T Reguladores/patología , Regulación hacia Arriba/inmunología
8.
PLoS One ; 10(9): e0138110, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26413862

RESUMEN

Current approaches to study transcriptional profiles post influenza infection typically rely on tissue sampling from one or two sites at a few time points, such as spleen and lung in murine models. In this study, we infected female C57/BL6 mice intranasally with mouse-adapted H3N2/Hong Kong/X31 avian influenza A virus, and then analyzed the gene expression profiles in four different compartments (blood, lung, mediastinal lymph nodes, and spleen) over 11 consecutive days post infection. These data were analyzed by an advanced statistical procedure based on ordinary differential equation (ODE) modeling. Vastly different lists of significant genes were identified by the same statistical procedure in each compartment. Only 11 of them are significant in all four compartments. We classified significant genes in each compartment into co-expressed modules based on temporal expression patterns. We then performed functional enrichment analysis on these co-expression modules and identified significant pathway and functional motifs. Finally, we used an ODE based model to reconstruct gene regulatory network (GRN) for each compartment and studied their network properties.


Asunto(s)
Redes Reguladoras de Genes , Inmunidad/genética , Virus de la Influenza A/fisiología , Especificidad de Órganos/genética , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/inmunología , Inmunidad Adaptativa/genética , Animales , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Inmunidad Innata/genética , Pulmón/metabolismo , Ganglios Linfáticos/metabolismo , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/virología , Factores de Tiempo , Transcriptoma/genética
9.
PLoS One ; 10(7): e0133791, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26208102

RESUMEN

Dysregulation of hematopoietic stem cell (HSC) signaling can contribute to the development of diseases of the blood system. Lack of aryl hydrocarbon receptor (AhR) has been associated with alterations in gene expression related to HSC function and the subsequent development of a myeloproliferative disorder in aging female mice. We sorted the most primitive population of HSCs with the highest stem cell potential (Long-term, or LT-HSCs) from 18-month-old AhR-null-allele (AhR-KO) and WT mice and analyzed gene expression using microarray to determine alterations in gene expression and cell signaling networks in HSCs that could potentially contribute to the aging phenotype of AhR-KO mice. Comparisons with previous array data from 8-week old mice indicated that aging alone is sufficient to alter gene expression. In addition, a significant number of gene expression differences were observed in aged LT-HSCs that are dependent on both aging and lack of AhR. Pathway analysis of these genes revealed networks related to hematopoietic stem cell activity or function. qPCR was used to confirm the differential expression of a subset of these genes, focusing on genes that may represent novel AhR targets due to the presence of a putative AhR binding site in their upstream regulatory region. We verified differential expression of PDGF-D, Smo, Wdfy1, Zbtb37 and Zfp382. Pathway analysis of this subset of genes revealed overlap between cellular functions of the novel AhR targets and AhR itself. Lentiviral-mediated knockdown of AhR in lineage-negative hematopoietic cells was sufficient to induce changes in all five of the candidate AhR targets identified. Taken together, these data suggest a role for AhR in HSC functional regulation, and identify novel HSC AhR target genes that may contribute to the phenotypes observed in AhR-KO mice.


Asunto(s)
Envejecimiento/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Células Madre Hematopoyéticas/metabolismo , Receptores de Hidrocarburo de Aril/genética , Transcriptoma , Animales , Análisis por Conglomerados , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Redes Reguladoras de Genes , Ratones , Ratones Noqueados , Fenotipo , Reproducibilidad de los Resultados , Transducción de Señal
10.
J Clin Invest ; 124(7): 3200-14, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24892805

RESUMEN

NOTCH-dependent signaling pathways are critical for normal bone remodeling; however, it is unclear if dysfunctional NOTCH activation contributes to inflammation-mediated bone loss, as observed in rheumatoid arthritis (RA) patients. We performed RNA sequencing and pathway analyses in mesenchymal stem cells (MSCs) isolated from transgenic TNF-expressing mice, a model of RA, to identify pathways responsible for decreased osteoblast differentiation. 53 pathways were dysregulated in MSCs from RA mice, among which expression of genes encoding NOTCH pathway members and members of the noncanonical NF-κB pathway were markedly elevated. Administration of NOTCH inhibitors to RA mice prevented bone loss and osteoblast inhibition, and CFU-fibroblasts from RA mice treated with NOTCH inhibitors formed more new bone in recipient mice with tibial defects. Overexpression of the noncanonical NF-κB subunit p52 and RELB in a murine pluripotent stem cell line increased NOTCH intracellular domain-dependent (NICD-dependent) activation of an RBPjκ reporter and levels of the transcription factor HES1. TNF promoted p52/RELB binding to NICD, which enhanced binding at the RBPjκ site within the Hes1 promoter. Furthermore, MSC-enriched cells from RA patients exhibited elevated levels of HES1, p52, and RELB. Together, these data indicate that persistent NOTCH activation in MSCs contributes to decreased osteoblast differentiation associated with RA and suggest that NOTCH inhibitors could prevent inflammation-mediated bone loss.


Asunto(s)
Artritis Reumatoide/metabolismo , FN-kappa B/metabolismo , Osteoblastos/metabolismo , Receptores Notch/metabolismo , Animales , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/patología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Resorción Ósea/metabolismo , Resorción Ósea/patología , Resorción Ósea/prevención & control , Diferenciación Celular , Dipéptidos/farmacología , Modelos Animales de Enfermedad , Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , FN-kappa B/genética , Subunidad p52 de NF-kappa B/genética , Subunidad p52 de NF-kappa B/metabolismo , Osteoblastos/efectos de los fármacos , Osteoblastos/patología , Regiones Promotoras Genéticas , Receptores Notch/antagonistas & inhibidores , Receptores Notch/genética , Transducción de Señal , Factor de Transcripción HES-1 , Factor de Transcripción ReIB/genética , Factor de Transcripción ReIB/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
11.
Stem Cells Dev ; 23(2): 95-106, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24138668

RESUMEN

Loss of immune function and increased hematopoietic disease are among the most clinically significant consequences of aging. Hematopoietic stem cells (HSCs) from mice lacking aryl hydrocarbon receptor (AhR) have high rates of cell division. Studies were designed to test the hypothesis that aging AhR-null allele (AhR-KO) mice develop premature HSC exhaustion, and changes leading to hematological disease. Compared to wild-type, aging AhR-KO mice showed a decreased survival rate, splenomegaly, increased circulating white blood cells, hematopoietic cell accumulation in tissues, and anemia. Analysis of bone marrow indicated increased numbers of stem/progenitor and lineage-committed cells, but decreased erythroid progenitors. There was also decreased self-renewal capacity of HSCs determined by competitive repopulation and serial transplantation. HSCs also showed increased levels of reactive oxygen species (ROS), Ki-67, and γ-H2A.X, but decreased p16(Ink4a). Splenic cells from aging KO mice had abnormal expression of genes, including Gata-1, Sh2d3c, Gfi-1, p21, and c-myc, involved in trafficking and associated with leukemia. HSCs from AhR-KO mice had gene changes related to HSC maintenance and consistent with phenotype observed. The most prominent gene changes (overexpression of Srpk2, Creb1, Hes1, mtor, pdp1) have been associated with HSC hyperproliferation, leukemia, and accelerated aging. Pathway analyses also indicated an enrichment of genes associated with oxidative stress, acute myelogenous leukemia, aging, and heat shock response, and the ß-catenin/Wnt pathways. These data indicate that loss of AhR and associated changes in multiple signaling pathways promote premature HSC exhaustion and development of a myeloproliferative disorder. They also implicate a critical role of the AhR in the regulation of HSCs.


Asunto(s)
Envejecimiento/genética , Envejecimiento/inmunología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Células Madre Hematopoyéticas/citología , Trastornos Mieloproliferativos/genética , Receptores de Hidrocarburo de Aril/genética , Anemia/genética , Animales , Células de la Médula Ósea/citología , Movimiento Celular/genética , Movimiento Celular/inmunología , Proliferación Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Femenino , Expresión Génica/genética , Trasplante de Células Madre Hematopoyéticas , Histonas/biosíntesis , Antígeno Ki-67/biosíntesis , Recuento de Leucocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo/genética , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/genética , Esplenomegalia/genética , Tasa de Supervivencia
12.
Sci Rep ; 3: 2327, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23900141

RESUMEN

To identify sources of inter-subject variation in vaccine responses, we performed high-frequency sampling of human peripheral blood cells post-vaccination, followed by a novel systems biology analysis. Functional principal component analysis was used to examine time varying B cell vaccine responses. In subjects vaccinated within the previous three years, 90% of transcriptome variation was explained by a single subject-specific mathematical pattern. Within individual vaccine response patterns, a common subset of 742 genes was strongly correlated with migrating plasma cells. Of these, 366 genes were associated with human plasmablasts differentiating in vitro. Additionally, subject-specific temporal transcriptome patterns in peripheral blood mononuclear cells identified migration of myeloid/dendritic cell lineage cells one day after vaccination. Upstream analyses of transcriptome changes suggested both shared and subject-specific transcription groups underlying larger patterns. With robust statistical methods, time-varying response characteristics of individual subjects were effectively captured along with a shared plasma cell gene signature.


Asunto(s)
Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/inmunología , Transcriptoma/inmunología , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/inmunología , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Transcriptoma/efectos de los fármacos
13.
Am J Physiol Endocrinol Metab ; 304(10): E1042-52, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23531613

RESUMEN

Although catabolic signaling has a well-established role in muscle wasting during cancer cachexia, the suppression of anabolic signaling also warrants further investigation. In cachectic tumor-bearing mice, circulating IL-6 levels are associated with suppressed muscle protein synthesis and mTORC1 signaling. We have found AMPK and IGF-I/insulin signaling, two well-known regulators of the mammalian target of rapamycin (mTOR), are altered with the progression of cachexia. How IL-6 can induce suppression of mTORC1 signaling remains to be established. The purpose of this study was to examine mTOR complex 1 (mTORC1) activation and regulation by IL-6 during cancer cachexia. IL-6 effects on mTOR activation were examined in Apc(Min/+) mouse skeletal muscle and C2C12 myotubes. Systemic IL-6 overexpression in Apc(Min/+) mice produced a dose-dependent suppression of mTOR signaling that corresponded to induction of STAT3 and AMPK phosphorylation. This result was also evident in IL-6-treated myotubes. Basal mTOR activation and mTOR responsiveness to glucose administration were suppressed in cachectic skeletal muscle. However, insulin induction of mTOR activity was maintained in IL-6-treated myotubes. Whereas IL-6 suppression of myotube mTOR activity was rescued by AMPK inhibition, inhibition of STAT3 signaling was not sufficient to rescue IL-6 suppression of mTOR activity. Last, treadmill exercise training was able to prevent IL-6-induced inhibition of mTOR signaling in Apc(Min/+) mice independently of activated STAT. In conclusion, we report dose-dependent suppression of mTOR activity by IL-6 and suppressed mTOR responsiveness to glucose administration in Apc(Min/+) mice. IL-6 suppression of mTOR activity was dependent on AMPK activation and independent of STAT signaling in myotubes.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Caquexia/metabolismo , Interleucina-6/metabolismo , Músculo Esquelético/metabolismo , Neoplasias Experimentales/metabolismo , Proteínas/metabolismo , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/genética , Aminoimidazol Carboxamida/farmacología , Animales , Western Blotting , Caquexia/enzimología , Interleucina-6/sangre , Interleucina-6/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Complejos Multiproteicos , Fibras Musculares Esqueléticas/enzimología , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/enzimología , Neoplasias Experimentales/enzimología , Fosforilación , Condicionamiento Físico Animal/fisiología , Proteínas/antagonistas & inhibidores , Proteínas/genética , Pirazoles/farmacología , Pirimidinas/farmacología , ARN Mensajero/química , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Recombinantes/farmacología , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR
14.
Mol Cell Endocrinol ; 365(2): 174-86, 2013 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-23116773

RESUMEN

Low endogenous testosterone production, known as hypogonadism is commonly associated with conditions inducing muscle wasting. Akt signaling can control skeletal muscle mass through mTOR regulation of protein synthesis and FoxO regulation of protein degradation, and this pathway has been previously identified as a target of androgen signaling. However, the testosterone sensitivity of Akt/mTOR signaling requires further understanding in order to grasp the significance of varied testosterone levels seen with wasting disease on muscle protein turnover regulation. Therefore, the purpose of this study is to determine the effect of androgen availability on muscle Akt/mTORC1/FoxO3a regulation in skeletal muscle and cultured C(2)C(12) myotubes. C57BL/6 mice were either castrated for 42 days or castrated and treated with the nandrolone decanoate (ND) (6 mg/kg bw/wk). Testosterone loss (TL) significantly decreased volitional grip strength, body weight, and gastrocnemius (GAS) muscle mass, and ND reversed these changes. Related to muscle mass regulation, TL decreased muscle IGF-1 mRNA, the rate of myofibrillar protein synthesis, Akt phosphorylation, and the phosphorylation of Akt targets, GSK3ß, PRAS40 and FoxO3a. TL induced expression of FoxO transcriptional targets, MuRF1, atrogin1 and REDD1. Muscle AMPK and raptor phosphorylation, mTOR inhibitors, were not altered by low testosterone. ND restored IGF-1 expression and Akt/mTORC1 signaling while repressing expression of FoxO transcriptional targets. Testosterone (T) sensitivity of Akt/mTORC1 signaling was examined in C(2)C(12) myotubes, and mTOR phosphorylation was induced independent of Akt activation at low T concentrations, while a higher T concentration was required to activate Akt signaling. Interestingly, low concentration T was sufficient to amplify myotube mTOR and Akt signaling after 24 h of T withdrawal, demonstrating the potential in cultured myotubes for a T initiated positive feedback mechanism to amplify Akt/mTOR signaling. In summary, androgen withdrawal decreases muscle myofibrillar protein synthesis through Akt/mTORC1 signaling, which is independent of AMPK activation, and readily reversible by anabolic steroid administration. Acute Akt activation in C(2)C(12) myotubes is sensitive to a high concentration of testosterone, and low concentrations of testosterone can activate mTOR signaling independent of Akt.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Testosterona/fisiología , Adenilato Quinasa/metabolismo , Andrógenos/farmacología , Animales , Línea Celular , Activación Enzimática , Proteína Forkhead Box O3 , Expresión Génica , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Complejos Multiproteicos , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/fisiología , Fuerza Muscular , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiología , Nandrolona/análogos & derivados , Nandrolona/farmacología , Nandrolona Decanoato , Orquiectomía , Fosforilación , Procesamiento Proteico-Postraduccional , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR , Activación Transcripcional
15.
Am J Pathol ; 181(2): 441-51, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22677423

RESUMEN

Oxygen exposure in premature infants is a major risk factor for bronchopulmonary dysplasia and can impair the host response to respiratory viral infections later in life. Similarly, adult mice exposed to hyperoxia as neonates display alveolar simplification associated with a reduced number of alveolar epithelial type II cells and exhibit persistent inflammation, fibrosis, and mortality when infected with influenza A virus. Because type II cells participate in innate immunity and alveolar repair, their loss may contribute to oxygen-mediated sensitivity to viral infection. A genomewide screening of type II cells identified eosinophil-associated RNase 1 (Ear1). Ear1 was also detected in airway epithelium and was reduced in lungs of mice exposed to neonatal hyperoxia. Electroporation-mediated gene delivery of Ear1 to the lung before infection successfully reduced viral replication and leukocyte recruitment during infection. It also diminished the enhanced morbidity and mortality attributed to neonatal hyperoxia. These findings demonstrate that novel epithelial expression of Ear1 functions to limit influenza A virus infection, and its loss contributes to oxygen-associated epithelial injury and fibrosis after infection. People born prematurely may have defects in epithelial innate immunity that increase their risk for respiratory viral infections.


Asunto(s)
Neurotoxina Derivada del Eosinófilo/metabolismo , Epitelio/metabolismo , Virus de la Influenza A/fisiología , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Oxígeno/farmacología , Ribonucleasas/metabolismo , Envejecimiento/patología , Aire , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Animales , Animales Recién Nacidos , Electroporación , Epitelio/efectos de los fármacos , Epitelio/patología , Epitelio/virología , Femenino , Técnicas de Transferencia de Gen , Hiperoxia/complicaciones , Hiperoxia/patología , Hiperoxia/virología , Virus de la Influenza A/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/prevención & control
16.
PLoS One ; 6(9): e24650, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21949739

RESUMEN

Muscle wasting that occurs with cancer cachexia is caused by an imbalance in the rates of muscle protein synthesis and degradation. The Apc(Min/+) mouse is a model of colorectal cancer that develops cachexia that is dependent on circulating IL-6. However, the IL-6 regulation of muscle protein turnover during the initiation and progression of cachexia in the Apc(Min/+) mouse is not known. Cachexia progression was studied in Apc(Min/+) mice that were either weight stable (WS) or had initial (≤5%), intermediate (6-19%), or extreme (≥20%) body weight loss. The initiation of cachexia reduced %MPS 19% and a further ∼50% with additional weight loss. Muscle IGF-1 mRNA expression and mTOR targets were suppressed with the progression of body weight loss, while muscle AMPK phosphorylation (Thr 172), AMPK activity, and raptor phosphorylation (Ser 792) were not increased with the initiation of weight loss, but were induced as cachexia progressed. ATP dependent protein degradation increased during the initiation and progression of cachexia. However, ATP independent protein degradation was not increased until cachexia had progressed beyond the initial phase. IL-6 receptor antibody administration prevented body weight loss and suppressed muscle protein degradation, without any effect on muscle %MPS or IGF-1 associated signaling. In summary, the %MPS reduction during the initiation of cachexia is associated with IGF-1/mTOR signaling repression, while muscle AMPK activation and activation of ATP independent protein degradation occur later in the progression of cachexia. IL-6 receptor antibody treatment blocked cachexia progression through the suppression of muscle protein degradation, while not rescuing the suppression of muscle protein synthesis. Attenuation of IL-6 signaling was effective in blocking the progression of cachexia, but not sufficient to reverse the process.


Asunto(s)
Caquexia/complicaciones , Caquexia/patología , Progresión de la Enfermedad , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Neoplasias/complicaciones , Neoplasias/patología , Proteínas Quinasas Activadas por AMP/metabolismo , Adenosina Trifosfato/metabolismo , Adiposidad , Animales , Peso Corporal , Caquexia/sangre , Activación Enzimática , Inflamación/complicaciones , Inflamación/patología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Interleucina-6/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Músculo Esquelético/enzimología , Músculo Esquelético/patología , Miofibrillas/metabolismo , Neoplasias/sangre , Tamaño de los Órganos , Fosforilación , Biosíntesis de Proteínas , Proteolisis , Transducción de Señal , Serina-Treonina Quinasas TOR
17.
Toxicol Sci ; 114(1): 90-100, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19933214

RESUMEN

The aryl hydrocarbon receptor (AhR), a ligand-dependent transcription factor, mediates toxicity of several classes of xenobiotics and also has important physiological roles in differentiation, reproduction, and immunity, although the endogenous ligand(s) mediating these functions is/are as yet unidentified. One candidate endogenous ligand, 2-(1'H-indolo-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE), is a potent AhR agonist in vitro, activates the murine AhR in vivo, but does not induce toxicity. We hypothesized that ITE and the toxic ligand, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), may modify transcription of different sets of genes to account for their different toxicity. To test this hypothesis, primary mouse lung fibroblasts were exposed to 0.5muM ITE, 0.2nM TCDD, or vehicle for 4 h, and total gene expression was evaluated using microarrays. After this short-term and low-dose treatment, several hundred genes were changed significantly, and the response to ITE and TCDD was remarkably similar, both qualitatively and quantitatively. Induced gene sets included the expected battery of AhR-dependent xenobiotic-metabolizing enzymes, as well as several sets that reflect the inflammatory role of lung fibroblasts. Real time quantitative RT-qPCR assay of several selected genes confirmed these microarray data and further suggested that there may be kinetic differences in expression between ligands. These data suggest that ITE and TCDD elicit an analogous change in AhR conformation such that the initial transcription response is the same. Furthermore, if the difference in toxicity between TCDD and ITE is mediated by differences in gene expression, then it is likely that secondary changes enabled by the persistent TCDD, but not by the shorter lived ITE, are responsible.


Asunto(s)
Fibroblastos/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Indoles/toxicidad , Pulmón/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Receptores de Hidrocarburo de Aril/metabolismo , Tiazoles/toxicidad , Animales , Relación Dosis-Respuesta a Droga , Fibroblastos/metabolismo , Ligandos , Pulmón/citología , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores de Hidrocarburo de Aril/genética
19.
Am J Physiol Cell Physiol ; 292(4): C1291-7, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17135300

RESUMEN

Transmembrane chloride ion conductance in skeletal muscle increases during early postnatal development. A transgenic mouse model of myotonic dystrophy type 1 (DM1) displays decreased sarcolemmal chloride conductance. Both effects result from modulation of chloride channel 1 (CLCN1) expression, but the respective contributions of transcriptional vs. posttranscriptional regulation are unknown. Here we show that alternative splicing of CLCN1 undergoes a physiological splicing transition during the first 3 wk of postnatal life in mice. During this interval, there is a switch to production of CLCN1 splice products having an intact reading frame, an upregulation of CLCN1 mRNA encoding full-length channel protein, and an increase of CLCN1 function, as determined by patch-clamp analysis of single muscle fibers. In a transgenic mouse model of DM1, however, the splicing transition does not occur, CLCN1 channel function remains low throughout the postnatal interval, and muscle fibers display myotonic discharges. Thus alternative splicing is a posttranscriptional mechanism regulating chloride conductance during muscle development, and the chloride channelopathy in a transgenic mouse model of DM1 results from a failure to execute a splicing transition for CLCN1.


Asunto(s)
Canalopatías/metabolismo , Canales de Cloruro/fisiología , Músculo Esquelético/metabolismo , Distrofia Miotónica/metabolismo , Procesamiento Postranscripcional del ARN , Empalme Alternativo , Animales , Canalopatías/genética , Canales de Cloruro/biosíntesis , Canales de Cloruro/genética , Técnicas In Vitro , Activación del Canal Iónico , Ratones , Ratones Transgénicos , Desarrollo de Músculos , Fibras Musculares Esqueléticas/fisiología , Músculo Esquelético/crecimiento & desarrollo , Distrofia Miotónica/genética , Técnicas de Placa-Clamp , Isoformas de Proteínas/biosíntesis , ARN Mensajero/biosíntesis
20.
Am J Physiol Endocrinol Metab ; 290(1): E67-E77, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16105859

RESUMEN

To assess mechanisms for postprandial hyperglycemia, we used a triple-isotope technique ([\3-(3)H]glucose and [(14)C]bicarbonate and oral [6,6-dideutero]glucose iv) and indirect calorimetry to compare components of glucose release and pathways for glucose disposal in 26 subjects with type 2 diabetes and 15 age-, weight-, and sex-matched normal volunteers after a standard meal. The results were as follows: 1) diabetic subjects had greater postprandial glucose release (P<0.001) because of both increased endogenous and meal-glucose release; 2) the greater endogenous glucose release (P<0.001) was due to increased gluconeogenesis (P<0.001) and glycogenolysis (P=0.01); 3) overall tissue glucose uptake, glycolysis, and storage were comparable in both groups (P>0.3); 4) glucose clearance (P<0.001) and oxidation (P=0.004) were reduced, whereas nonoxidative glycolysis was increased (P=0.04); and 5) net splanchnic glucose storage was reduced by approximately 45% (P=0.008) because of increased glycogen cycling (P=0.03). Thus in type 2 diabetes, postprandial hyperglycemia is primarily due to increased glucose release; hyperglycemia overcomes the effects of impaired insulin secretion and sensitivity on glucose transport, but intracellular defects persist so that pathways of glucose metabolism are abnormal and glucose is shunted away from normal sites of storage (e.g., liver and muscle) into other tissues.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Glucosa/metabolismo , Periodo Posprandial/fisiología , Alanina/sangre , Glucemia/metabolismo , Dióxido de Carbono/metabolismo , Diabetes Mellitus Tipo 2/sangre , Ácidos Grasos no Esterificados/sangre , Femenino , Glucagón/sangre , Gluconeogénesis/fisiología , Glicerol/sangre , Glucogenólisis/fisiología , Glucólisis/fisiología , Humanos , Hiperglucemia/sangre , Hiperglucemia/metabolismo , Insulina/sangre , Ácido Láctico/sangre , Masculino , Persona de Mediana Edad , Oxidación-Reducción , Vísceras/metabolismo , Agua/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...