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










Base de datos
Intervalo de año de publicación
1.
Mol Cancer Res ; 20(8): 1233-1246, 2022 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-35533313

RESUMEN

Coevolution of tumor cells and adjacent stromal elements is a key feature during tumor progression; however, the precise regulatory mechanisms during this process remain unknown. Here, we show stromal p53 loss enhances oncogenic KrasG12D, but not ErbB2, driven tumorigenesis in murine mammary epithelia. Stroma-specific p53 deletion increases both epithelial and fibroblast proliferation in mammary glands bearing the KrasG12D oncogene in epithelia, while concurrently increasing DNA damage and/or DNA replication stress and decreasing apoptosis in the tumor cells proper. Normal epithelia was not affected by stromal p53 deletion. Tumors with p53-null stroma had a significant decrease in total, cytotoxic, and regulatory T cells; however, there was a significant increase in myeloid-derived suppressor cells, total macrophages, and M2-polarized tumor-associated macrophages, with no impact on angiogenesis or connective tissue deposition. Stroma-specific p53 deletion reprogrammed gene expression in both fibroblasts and adjacent epithelium, with p53 targets and chemokine receptors/chemokine signaling pathways in fibroblasts and DNA replication, DNA damage repair, and apoptosis in epithelia being the most significantly impacted biological processes. A gene cluster in p53-deficient mouse fibroblasts was negatively associated with patient survival when compared with two independent datasets. In summary, stroma-specific p53 loss promotes mammary tumorigenesis in an oncogene-specific manner, influences the tumor immune landscape, and ultimately impacts patient survival. IMPLICATIONS: Expression of the p53 tumor suppressor in breast cancer tumor stroma regulates tumorigenesis in an oncogene-specific manner, influences the tumor immune landscape, and ultimately impacts patient survival.


Asunto(s)
Neoplasias de la Mama , Oncogenes , Proteína p53 Supresora de Tumor , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Carcinogénesis , Tejido Conectivo/metabolismo , Ratones , Proteínas Proto-Oncogénicas p21(ras) , Células del Estroma/patología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
2.
Cancer Metab ; 9(1): 39, 2021 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-34749809

RESUMEN

BACKGROUND: Pyruvate dehydrogenase complex (PDC) plays a central role in carbohydrate metabolism, linking cytoplasmic glycolysis to the mitochondrial tricarboxylic acid (TCA) cycle. PDC is a conserved E1-E2-E3 dehydrogenase with a PDHA1 and PDHB heterotetramer functioning as the E1 subunit. PDHA1 contains three serine residues that can be reversibly phosphorylated by a dedicated family of four inhibitory pyruvate dehydrogenase kinases (PDHK1-4) and two reactivating phosphatases (PDP1, 2). Hypoxia induces the expression of PDHK1 and PDHK3 and hyperphosphorylates PDHA1. The role of PDC in metabolic reprogramming and tumor progression appears to be for the integration of oncogenic and environmental signals which supports tumor growth. METHODS: To isolate the function of the serine-dependent regulation of PDC, we engineered MiaPaca2 cells to express PDHA1 protein with either intact serines at positions 232, 293, and 300 or all the combinations of non-phosphorylatable alanine substitution mutations. These lines were compared in vitro for biochemical response to hypoxia by western blot, metabolic activity by biochemical assay and Seahorse XF flux analysis, and growth in media with reduced exogenous metabolites. The lines were also tested for growth in vivo after orthotopic injection into the pancreata of immune-deficient mice. RESULTS: In this family of cells with non-phosphorylatable PDHA1, we found reduced hypoxic phosphorylation of PDHA1, decreased PDH enzymatic activity in normoxia and hypoxia, decreased mitochondrial function by Seahorse flux assay, reduced in vitro growth of cells in media depleted of lipids, and reduced growth of tumors after orthotopic transplantation of cells into the pancreata of immune-deficient mice. CONCLUSIONS: We found that any substitution of alanine for serine at regulatory sites generated a hypomorphic PDC. However, the reduced PDC activity was insensitive to further reduction in hypoxia. These cells had a very modest reduction of growth in vitro, but failed to grow as tumors indicating that dynamic PDC adaptation to microenvironmental conditions is necessary to support pancreatic cancer growth in vivo.

3.
Cancers (Basel) ; 13(7)2021 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-33916656

RESUMEN

BACKGROUND: Hypoxia is found in many solid tumors and is associated with increased disease aggressiveness and resistance to therapy. Reducing oxygen demand by targeting mitochondrial oxidative metabolism is an emerging concept in translational cancer research aimed at reducing hypoxia. We have shown that the U.S. Food and Drug Administration (FDA)-approved drug papaverine and its novel derivative SMV-32 are potent mitochondrial complex I inhibitors. METHODS: We used a dynamic in vivo luciferase reporter system, pODD-Luc, to evaluate the impact of pharmacological manipulation of mitochondrial metabolism on the levels of tumor hypoxia in transplanted mouse tumors. We also imaged canine patients with blood oxygen level-dependent (BOLD) MRI at baseline and one hour after a dose of 1 or 2 mg/kg papaverine. RESULTS: We showed that the pharmacological suppression of mitochondrial oxygen consumption (OCR) in tumor-bearing mice increases tumor oxygenation, while the stimulation of mitochondrial OCR decreases tumor oxygenation. In parallel experiments in a small series of spontaneous canine sarcomas treated at The Ohio State University (OSU) Veterinary Medical Center, we observed a significant increase in BOLD signals indicative of an increase in tumor oxygenation of up to 10-50 mm HgO2. CONCLUSION: In both transplanted murine tumors and spontaneous canine tumors we found that decreasing mitochondrial metabolism can decrease tumor hypoxia, potentially offering a therapeutic advantage.

4.
Mol Cancer Res ; 17(10): 2089-2101, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31308147

RESUMEN

Accumulation of lipid droplets has been observed in an increasing range of tumors. However, the molecular determinants of this phenotype and the impact of the tumor microenvironment on lipid droplet dynamics are not well defined. The hypoxia-inducible and lipid droplet associated protein HILPDA is known to regulate lipid storage and physiologic responses to feeding conditions in mice, and was recently shown to promote hypoxic lipid droplet formation through inhibition of the rate-limiting lipase adipose triglyceride lipase (ATGL). Here, we identify fatty acid loading and nutrient deprivation-induced autophagy as stimuli of HILPDA-dependent lipid droplet growth. Using mouse embryonic fibroblasts and human tumor cells, we found that genetic ablation of HILPDA compromised hypoxia-fatty acid- and starvation-induced lipid droplet formation and triglyceride storage. Nutrient deprivation upregulated HILPDA protein posttranscriptionally by a mechanism requiring autophagic flux and lipid droplet turnover, independent of HIF1 transactivation. Mechanistically, loss of HILPDA led to elevated lipolysis, which could be corrected by inhibition of ATGL. Lipidomic analysis revealed not only quantitative but also qualitative differences in the glycerolipid and phospholipid profile of HILPDA wild-type and knockout cells, indicating additional HILPDA functions affecting lipid metabolism. Deletion studies of HILPDA mutants identified the N-terminal hydrophobic domain as sufficient for targeting to lipid droplets and restoration of triglyceride storage. In vivo, HILPDA-ablated cells showed decreased intratumoral triglyceride levels and impaired xenograft tumor growth associated with elevated levels of apoptosis. IMPLICATIONS: Tumor microenvironmental stresses induce changes in lipid droplet dynamics via HILPDA. Regulation of triglyceride hydrolysis is crucial for cell homeostasis and tumor growth.


Asunto(s)
Gotas Lipídicas/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Animales , Femenino , Células HCT116 , Xenoinjertos , Humanos , Lipasa/metabolismo , Metabolismo de los Lípidos , Ratones , Ratones Noqueados , Ratones Desnudos , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Microambiente Tumoral , Regulación hacia Arriba
5.
Life Sci Alliance ; 1(5): e201800190, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30456390

RESUMEN

The contribution of the tumor microenvironment to pancreatic ductal adenocarcinoma (PDAC) development is currently unclear. We therefore examined the consequences of disrupting paracrine Hedgehog (HH) signaling in PDAC stroma. Herein, we show that ablation of the key HH signaling gene Smoothened (Smo) in stromal fibroblasts led to increased proliferation of pancreatic tumor cells. Furthermore, Smo deletion resulted in proteasomal degradation of the tumor suppressor PTEN and activation of oncogenic protein kinase B (AKT) in fibroblasts. An unbiased proteomic screen identified RNF5 as a novel E3 ubiquitin ligase responsible for degradation of phosphatase and tensin homolog (PTEN) in Smo-null fibroblasts. Ring Finger Protein 5 (Rnf5) knockdown or pharmacological inhibition of glycogen synthase kinase 3ß (GSKß), the kinase that marks PTEN for ubiquitination, rescued PTEN levels and reversed the oncogenic phenotype, identifying a new node of PTEN regulation. In PDAC patients, low stromal PTEN correlated with reduced overall survival. Mechanistically, PTEN loss decreased hydraulic permeability of the extracellular matrix, which was reversed by hyaluronidase treatment. These results define non-cell autonomous tumor-promoting mechanisms activated by disruption of the HH/PTEN axis and identifies new targets for restoring stromal tumor-suppressive functions.

6.
Proc Natl Acad Sci U S A ; 115(42): 10756-10761, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30201710

RESUMEN

Tumor hypoxia reduces the effectiveness of radiation therapy by limiting the biologically effective dose. An acute increase in tumor oxygenation before radiation treatment should therefore significantly improve the tumor cell kill after radiation. Efforts to increase oxygen delivery to the tumor have not shown positive clinical results. Here we show that targeting mitochondrial respiration results in a significant reduction of the tumor cells' demand for oxygen, leading to increased tumor oxygenation and radiation response. We identified an activity of the FDA-approved drug papaverine as an inhibitor of mitochondrial complex I. We also provide genetic evidence that papaverine's complex I inhibition is directly responsible for increased oxygenation and enhanced radiation response. Furthermore, we describe derivatives of papaverine that have the potential to become clinical radiosensitizers with potentially fewer side effects. Importantly, this radiosensitizing strategy will not sensitize well-oxygenated normal tissue, thereby increasing the therapeutic index of radiotherapy.


Asunto(s)
Hipoxia de la Célula/efectos de los fármacos , Neoplasias Pulmonares/radioterapia , Mitocondrias/efectos de los fármacos , NADH Deshidrogenasa/antagonistas & inhibidores , Oxígeno/metabolismo , Papaverina/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Animales , Sistemas CRISPR-Cas , Hipoxia de la Célula/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Complejo I de Transporte de Electrón , Femenino , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Mitocondrias/efectos de la radiación , NADH Deshidrogenasa/genética , Inhibidores de Fosfodiesterasa/farmacología , Tolerancia a Radiación , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
PLoS One ; 12(9): e0184984, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28934293

RESUMEN

The contribution of the tumor microenvironment to the development of pancreatic adenocarcinoma (PDAC) is unclear. The LSL-KrasG12D/+;LSL-p53R172H/+;Pdx-1-Cre (KPC) tumor model, which is widely utilized to faithfully recapitulate human pancreatic cancer, depends on Cre-mediated recombination in the epithelial lineage to drive tumorigenesis. Therefore, specific Cre-loxP recombination in stromal cells cannot be applied in this model, limiting the in vivo investigation of stromal genetics in tumor initiation and progression. To address this issue, we generated a new Pdx1FlpO knock-in mouse line, which represents the first mouse model to physiologically express FlpO recombinase in pancreatic epithelial cells. This mouse specifically recombines Frt loci in pancreatic epithelial cells, including acinar, ductal, and islet cells. When combined with the Frt-STOP-Frt KrasG12D and p53Frt mouse lines, simultaneous Pdx1FlpO activation of mutant Kras and deletion of p53 results in the spectrum of pathologic changes seen in PDAC, including PanIN lesions and ductal carcinoma. Combination of this KPF mouse model with any stroma-specific Cre can be used to conditionally modify target genes of interest. This will provide an excellent in vivo tool to study the roles of genes in different cell types and multiple cell compartments within the pancreatic tumor microenvironment.


Asunto(s)
Transformación Celular Neoplásica/patología , ADN Nucleotidiltransferasas/metabolismo , Modelos Animales de Enfermedad , Proteínas de Homeodominio/fisiología , Neoplasias Pancreáticas/patología , Transactivadores/fisiología , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , ADN Nucleotidiltransferasas/genética , Progresión de la Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Transducción de Señal , Microambiente Tumoral , Proteína p53 Supresora de Tumor/genética
8.
J Endocrinol ; 235(1): 27-38, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28739822

RESUMEN

Hypoxia-inducible lipid droplet-associated protein (HILPDA) has been shown to localize to lipid droplets in nutrient-responsive cell types such as hepatocytes and adipocytes. However, its role in the control of whole-body homeostasis is not known. We sought to measure cell-intrinsic and systemic stress responses in a mouse strain harboring whole-body Hilpda deficiency. We generated a genetically engineered mouse model of whole-body HILPDA deficiency by replacing the coding Hilpda exon with luciferase. We subjected the knockout animals to environmental stresses and measured whole-animal metabolic and behavioral parameters. Brown adipocyte precursors were isolated and differentiated in vitro to quantify the impact of HILPDA ablation in lipid storage and mobilization in these cells. HILPDA-knockout animals are viable and fertile, but show reduced ambulatory activity and oxygen consumption at regular housing conditions. Acclimatization at thermoneutral conditions abolished the phenotypic differences observed at 22°C. When fasted, HILPDA KO mice are unable to maintain body temperature and become hypothermic at 22°C, without apparent abnormalities in blood chemistry parameters or tissue triglyceride content. HILPDA expression was upregulated during adipocyte differentiation and activation in vitro; however, it was not required for lipid droplet formation in brown adipocytes. We conclude that HILPDA is necessary for efficient fuel utilization suggesting a homeostatic role for Hilpda in sub-optimal environments.


Asunto(s)
Regulación de la Temperatura Corporal , Proteínas de Unión al ADN/metabolismo , Ayuno/fisiología , Adipocitos/metabolismo , Animales , Proteínas de Unión al ADN/genética , Femenino , Ratones , Ratones Noqueados , Estrés Fisiológico , Triglicéridos/metabolismo
9.
Neoplasia ; 18(9): 541-52, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27659014

RESUMEN

Preclinical studies have suggested that the pancreatic tumor microenvironment both inhibits and promotes tumor development and growth. Here we establish the role of stromal fibroblasts during acinar-to-ductal metaplasia (ADM), an initiating event in pancreatic cancer formation. The transcription factor V-Ets avian erythroblastosis virus E26 oncogene homolog 2 (ETS2) was elevated in smooth muscle actin-positive fibroblasts in the stroma of pancreatic ductal adenocarcinoma (PDAC) patient tissue samples relative to normal pancreatic controls. LSL-Kras(G12D/+); LSL-Trp53(R172H/+); Pdx-1-Cre (KPC) mice showed that ETS2 expression initially increased in fibroblasts during ADM and remained elevated through progression to PDAC. Conditional ablation of Ets-2 in pancreatic fibroblasts in a Kras(G12D)-driven mouse ADM model decreased the amount of ADM events. ADMs from fibroblast Ets-2-deleted animals had reduced epithelial cell proliferation and increased apoptosis. Surprisingly, fibroblast Ets-2 deletion significantly altered immune cell infiltration into the stroma, with an increased CD8+ T-cell population, and decreased presence of regulatory T cells (Tregs), myeloid-derived suppressor cells, and mature macrophages. The mechanism involved ETS2-dependent chemokine ligand production in fibroblasts. ETS2 directly bound to regulatory sequences for Ccl3, Ccl4, Cxcl4, Cxcl5, and Cxcl10, a group of chemokines that act as potent mediators of immune cell recruitment. These results suggest an unappreciated role for ETS2 in fibroblasts in establishing an immune-suppressive microenvironment in response to oncogenic Kras(G12D) signaling during the initial stages of tumor development.


Asunto(s)
Células Acinares/metabolismo , Transformación Celular Neoplásica/metabolismo , Quimiocinas/biosíntesis , Quimiotaxis de Leucocito , Conductos Pancreáticos/metabolismo , Proteína Proto-Oncogénica c-ets-2/metabolismo , Células del Estroma/metabolismo , Células Acinares/patología , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Quimiotaxis de Leucocito/inmunología , Colágeno/metabolismo , Fibroblastos/metabolismo , Eliminación de Gen , Expresión Génica , Humanos , Inmunohistoquímica , Metaplasia , Ratones , Ratones Noqueados , Conductos Pancreáticos/inmunología , Conductos Pancreáticos/patología , Fenotipo , Proteína Proto-Oncogénica c-ets-2/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Transducción de Señal
10.
Genes Dev ; 30(17): 1943-55, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27633013

RESUMEN

The contribution of the microenvironment to pancreatic acinar-to-ductal metaplasia (ADM), a preneoplastic transition in oncogenic Kras-driven pancreatic cancer progression, is currently unclear. Here we show that disruption of paracrine Hedgehog signaling via genetic ablation of Smoothened (Smo) in stromal fibroblasts in a Kras(G12D) mouse model increased ADM. Smo-deleted fibroblasts had higher expression of transforming growth factor-α (Tgfa) mRNA and secreted higher levels of TGFα, leading to activation of EGFR signaling in acinar cells and increased ADM. The mechanism involved activation of AKT and noncanonical activation of the GLI family transcription factor GLI2. GLI2 was phosphorylated at Ser230 in an AKT-dependent fashion and directly regulated Tgfa expression in fibroblasts lacking Smo Additionally, Smo-deleted fibroblasts stimulated the growth of Kras(G12D)/Tp53(R172H) pancreatic tumor cells in vivo and in vitro. These results define a non-cell-autonomous mechanism modulating Kras(G12D)-driven ADM that is balanced by cross-talk between Hedgehog/SMO and AKT/GLI2 pathways in stromal fibroblasts.


Asunto(s)
Carcinoma Ductal Pancreático , Metaplasia/genética , Metaplasia/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Receptor Smoothened/genética , Receptor Smoothened/metabolismo , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Proliferación Celular/genética , Células Cultivadas , Células Epiteliales/metabolismo , Receptores ErbB/metabolismo , Fibroblastos/citología , Fibroblastos/patología , Eliminación de Gen , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Endogámicos C57BL , Páncreas/patología , Transducción de Señal/genética , Factor de Crecimiento Transformador alfa/metabolismo , Células Tumorales Cultivadas , Proteína Gli2 con Dedos de Zinc
11.
Cancer Res ; 71(7): 2622-31, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21310824

RESUMEN

Regulation of diurnal and circadian rhythms and cell proliferation are coupled in all mammals, including humans. However, the molecular mechanisms by which diurnal and circadian rhythms regulate cell proliferation are relatively poorly understood. In this study, we report that tumor growth in nude rats bearing human steroid receptor-negative MCF-7 breast tumors can be significantly accelerated by exposing the rats to light at night (LAN). Under normal conditions of an alternating light/dark cycle, proliferating cell nuclear antigen (PCNA) levels in tumors were maximal in the early light phase but remained at very low levels throughout the daily 24-hour cycle period monitored. Surprisingly, PCNA was expressed in tumors continually at a high level throughout the entire 24-hour period in LAN-exposed nude rats. Daily fluctuations of Akt and mitogen activated protein kinase activation in tumors were also disrupted by LAN. These fluctuations did not track with PCNA changes, but we found that activation of the Akt stimulatory kinase phosphoinositide-dependent protein kinase 1 (PDK1) directly correlated with PCNA levels. Expression of insulin-like growth factor 1 receptor (IGF-1R), an upstream signaling molecule for PDK1, also correlated with fluctuations of PDK1/PCNA in the LAN group. In addition, circulating IGF-1 concentrations were elevated in LAN-exposed tumor-bearing nude rats. Finally, RNAi-mediated knockdown of PDK1 led to a reduction in PCNA expression and cell proliferation in vitro and tumor growth in vivo, indicating that PDK1 regulates breast cancer growth in a manner correlated with PCNA expression. Taken together, our findings demonstrate that LAN exposure can accelerate tumor growth in vivo, in part through continuous activation of IGF-1R/PDK1 signaling.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ritmo Circadiano/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor IGF Tipo 1/metabolismo , Animales , Procesos de Crecimiento Celular/fisiología , Procesos de Crecimiento Celular/efectos de la radiación , Línea Celular Tumoral , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Luz , Antígeno Nuclear de Célula en Proliferación/biosíntesis , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Ratas , Ratas Desnudas , Transducción de Señal/efectos de la radiación , Trasplante Heterólogo
12.
Comp Med ; 60(5): 348-56, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21262119

RESUMEN

Dark-phase light contamination can significantly disrupt chronobiologic rhythms, thereby potentially altering the endocrine physiology and metabolism of experimental animals and influencing the outcome of scientific investigations. We sought to determine whether exposure to low-level light contamination during the dark phase influenced the normally entrained circadian rhythms of various substances in plasma. Male Sprague-Dawley rats (n = 6 per group) were housed in photobiologic light-exposure chambers configured to create 1) a 12:12-h light:dark cycle without dark-phase light contamination (control condition; 123 µW/cm(2), lights on at 0600), 2) experimental exposure to a low level of light during the 12-h dark phase (with 0.02, 0.05, 0.06, or 0.08 µW/cm(2) light at night), or 3) constant bright light (123 µW/cm(2)). Dietary and water intakes were recorded daily. After 2 wk, rats underwent 6 low-volume blood draws at 4-h intervals (beginning at 0400) during both the light and dark phases. Circadian rhythms in dietary and water intake and levels of plasma total fatty acids and lipid fractions remained entrained during exposure to either control conditions or low-intensity light during the dark phase. However, these patterns were disrupted in rats exposed to constant bright light. Circadian patterns of plasma melatonin, glucose, lactic acid, and corticosterone were maintained in all rats except those exposed to constant bright light or the highest level of light during the dark phase. Therefore even minimal light contamination during the dark phase can disrupt normal circadian rhythms of endocrine metabolism and physiology and may alter the outcome of scientific investigations.


Asunto(s)
Ritmo Circadiano/efectos de la radiación , Luz , Fotoperiodo , Ratas/fisiología , Animales , Glucemia/metabolismo , Corticosterona/sangre , Sistema Endocrino/efectos de la radiación , Ácidos Grasos/sangre , Vivienda para Animales , Ciencia de los Animales de Laboratorio , Ácido Láctico/sangre , Masculino , Melatonina/sangre , Ratas/metabolismo , Ratas Sprague-Dawley
13.
Circulation ; 119(17): 2357-2366, 2009 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-19380620

RESUMEN

BACKGROUND: Recent studies have identified critical roles for microRNAs (miRNAs) in a variety of cellular processes, including regulation of cardiomyocyte death. However, the signature of miRNA expression and possible roles of miRNA in the ischemic heart have been less well studied. METHODS AND RESULTS: We performed miRNA arrays to detect the expression pattern of miRNAs in murine hearts subjected to ischemia/reperfusion (I/R) in vivo and ex vivo. Surprisingly, we found that only miR-320 expression was significantly decreased in the hearts on I/R in vivo and ex vivo. This was further confirmed by TaqMan real-time polymerase chain reaction. Gain-of-function and loss-of-function approaches were employed in cultured adult rat cardiomyocytes to investigate the functional roles of miR-320. Overexpression of miR-320 enhanced cardiomyocyte death and apoptosis, whereas knockdown was cytoprotective, on simulated I/R. Furthermore, transgenic mice with cardiac-specific overexpression of miR-320 revealed an increased extent of apoptosis and infarction size in the hearts on I/R in vivo and ex vivo relative to the wild-type controls. Conversely, in vivo treatment with antagomir-320 reduced infarction size relative to the administration of mutant antagomir-320 and saline controls. Using TargetScan software and proteomic analysis, we identified heat-shock protein 20 (Hsp20), a known cardioprotective protein, as an important candidate target for miR-320. This was validated experimentally by utilizing a luciferase/GFP reporter activity assay and examining the expression of Hsp20 on miR-320 overexpression and knockdown in cardiomyocytes. CONCLUSIONS: Our data demonstrate that miR-320 is involved in the regulation of I/R-induced cardiac injury and dysfunction via antithetical regulation of Hsp20. Thus, miR-320 may constitute a new therapeutic target for ischemic heart diseases.


Asunto(s)
Proteínas del Choque Térmico HSP20/genética , MicroARNs/fisiología , Daño por Reperfusión Miocárdica/genética , Animales , Cardiotónicos , Muerte Celular , Células Cultivadas , Perfilación de la Expresión Génica , Ratones , Ratones Transgénicos , MicroARNs/genética , Miocitos Cardíacos
14.
J Biol Chem ; 283(7): 3942-50, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18063572

RESUMEN

Vasculogenesis, angiogenesis, and maturation are three major phases of the development of blood vessels. Although many receptors required for blood vessel formation have been defined, the intracellular signal transduction pathways involved in vascular maturation remain unclear. KLF2(-/-) embryos fail to develop beyond 13.5 days because of a lack of blood vessel stabilization. The molecular mechanism of KLF2 function in embryonic vascular vessels is still largely unknown. Here we show a normal development pattern of endothelial cells in KLF2(-/-) embryos but a defect of smooth muscle cells at the dorsal side of the aorta. This phenotype results from arrested vascular maturation characterized by the failure of mural cells to migrate around endothelial cells. This migration defect is also observed when platelet-derived growth factor-B (PDGF) controlled migration is studied in murine embryonic fibroblast (MEF) cells from KLF2(-/-) animals. In addition, KLF2(-/-) MEFs exhibit a significant growth defect, indicating that KLF2 is required to maintain the viability of MEF cells. The PDGF signal is mediated through the Src signaling pathway, and a downstream target of KLF2 is sphingosine 1-phosphate receptor 1. These studies demonstrate that KLF2 is required for smooth muscle cell migration and elucidate a novel mechanism involving communication between PDGF and KLF2 in vascular maturation.


Asunto(s)
Vasos Sanguíneos/crecimiento & desarrollo , Movimiento Celular/fisiología , Factores de Transcripción de Tipo Kruppel/fisiología , Músculo Liso Vascular/crecimiento & desarrollo , Animales , Vasos Sanguíneos/citología , Ratones , Ratones Noqueados , Músculo Liso Vascular/citología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal
15.
Nature ; 442(7100): 299-302, 2006 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-16855590

RESUMEN

Mammalian Kruppel-like transcription factors are implicated in regulating terminal differentiation of several tissue types. Deficiency in Kruppel-like factor (KLF) 2 (also known as LKLF) leads to a massive loss of the peripheral T-cell pool, suggesting KLF2 regulates T-cell quiescence and survival. Here we show, however, that KLF2 is essential for T-cell trafficking. KLF2-deficient (Klf2-/-) thymocytes show impaired expression of several receptors required for thymocyte emigration and peripheral trafficking, including the sphingosine-1-phosphate (S1P) receptor S1P1, CD62L and beta7 integrin. Furthermore, KLF2 both binds and transactivates the promoter for S1P1--a receptor that is critical for thymocyte egress and recirculation through peripheral lymphoid organs. Our findings suggest that KLF2 serves to license mature T cells for trafficking from the thymus and recirculation through secondary lymphoid tissues.


Asunto(s)
Movimiento Celular , Factores de Transcripción de Tipo Kruppel/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo , Timo/citología , Traslado Adoptivo , Animales , Línea Celular Tumoral , Quimera/metabolismo , Feto , Humanos , Células Jurkat , Factores de Transcripción de Tipo Kruppel/deficiencia , Factores de Transcripción de Tipo Kruppel/genética , Hígado/embriología , Ratones , Regiones Promotoras Genéticas/genética , Receptores de Lisoesfingolípidos/genética , Linfocitos T/trasplante , Activación Transcripcional
16.
J Immunol ; 175(5): 3060-6, 2005 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16116194

RESUMEN

Ag presentation to T lymphocytes and subsequent activation are characterized by a cascade of signaling events, some of which result in the transcriptional activation of a diverse set of genes. An important example is the induction of the IL-2 gene, which is a critical event in the escalation of T cell activation. Previous studies have found that expression of Krüppel-like factor 2 (KLF2), a zinc finger transcription factor, is extinguished after T cell activation. However, the biological role of KLF2 during T cell activation is still unknown. In this study we found that KLF2 protein degradation is delayed, and KLF2 expression is up-regulated during the early stage of T cell activation in primary T cells. Within a few hours, this process is reversed, and KLF2 expression is turned off. Next, we found that the expression of KLF2 significantly increases IL-2 production 4-fold in activated T cells, resulting from activation of the IL-2 promoter. By narrowing down the 2.0-kb IL-2 promoter region, we found that the KLF2 responsive element in the IL-2 promoter is a CACCC element, the KLF consensus binding motif. Moreover, KLF2 binds to this promoter in vivo under different conditions. Our studies show that KLF2 regulates IL-2 promoter activity in the earliest stages of T cell activation, indicating that KLF2 may act as a novel immediate-early transcriptional factor to maximally prime T cell activation.


Asunto(s)
Regulación de la Expresión Génica , Interleucina-2/genética , Activación de Linfocitos , Linfocitos T/inmunología , Transactivadores/fisiología , Secuencia de Bases , ADN/biosíntesis , Humanos , Células Jurkat , Factores de Transcripción de Tipo Kruppel , Datos de Secuencia Molecular , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , Elementos de Respuesta/fisiología , Transducción de Señal
17.
Biochemistry ; 44(33): 11098-105, 2005 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-16101293

RESUMEN

Kruppel-like transcription factor 2 (KLF2), a critical gene for mouse embryogenesis, was recently identified as an inhibitor of adipogenesis. However, it is still unknown whether KLF2 is a natural repressor of adipocyte differentiation and if KLF2 affects the formation of preadipocytes. It may also be important for preadipocyte formation, as KLF2 is crucial for lung development and blood vessel formation. In this study, we show that differentiation of preadipocytes not only results in a concomitant decrease in the levels of KLF2 protein but also significantly reduces KLF2 promoter activity. We have generated tet-responsive lines of 3T3L1 that express physiological levels of KLF2 and show that reexpression of KLF2 prevents preadipocyte differentiation, thereby confirming the inhibition of adipogenesis by KLF2, partially via the restoration of Pref-1. In addition, we studied the contribution of KLF2-negative cells to the formation and subsequent differentiation of preadipocytes. We demonstrate that embryoid bodies derived from KLF2(-)(/)(-) ES cells can differentiate into adipocytes as evidenced by the accumulation of lipids and expression of several biochemical markers. Moreover, mouse embryonic fibroblasts (MEFs) derived from KLF2(-)(/)(-) mouse embryos differentiate efficiently into adipocytes. Interestingly, quantification of lipid accumulation in MEFs indicated that KLF2(-)(/)(-) cells are more prone to differentiate at the early stage of the process, suggesting that KLF2 is a natural repressor of differentiation in vivo. Taken together, these studies demonstrate that KLF2 does not affect the commitment of multipotent stem cells into the preadipocytic lineage but rather maintains their preadipocyte state and thereby negatively regulates their transition into adipocytes.


Asunto(s)
Adipocitos/fisiología , Diferenciación Celular/genética , Linaje de la Célula/genética , Expresión Génica , Células Madre Multipotentes/fisiología , Transactivadores/genética , Adipocitos/citología , Animales , Vasos Sanguíneos/embriología , Línea Celular , Factores de Transcripción de Tipo Kruppel , Metabolismo de los Lípidos , Pulmón/embriología , Ratones , Ratones Noqueados , Células Madre Multipotentes/citología , Neovascularización Fisiológica/genética , Transactivadores/metabolismo , Transfección
18.
J Dermatol ; 31(7): 535-9, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15492417

RESUMEN

We report a case of acrogeria. A 47-year-old Japanese man presented with micrognathism, thin lips, radial wrinkles around his month, atrophy of skin and subcutaneous tissue, and mottled hyperpigmentation on his extremities. A biopsy of the lesional skin showed flat epidermis and atrophy of the dermal layer. The in vitro life span of the patient's fibroblasts (18+/-2.2 PDL) was significantly shorter than that of control fibroblasts (42+/-3.5 PDL). The early-passage fibroblasts from the patient showed abnormal morphology which was also seen in the late-passage (in vitro aging) of normal fibroblasts. In northern blotting analysis of cultured dermal fibroblasts, mRNA levels of alpha1 (I) collagen and alpha1 (III) collagen were markedly reduced. These results revealed that patient fibroblasts might be in severe senescence in vitro and contribute to the phenotypes of this premature aging syndrome.


Asunto(s)
Colágeno/metabolismo , Progeria/genética , Progeria/patología , Biopsia con Aguja , Northern Blotting , Células Cultivadas , Colágeno/clasificación , Fibroblastos/fisiología , Estudios de Seguimiento , Regulación de la Expresión Génica , Humanos , Inmunohistoquímica , Japón , Masculino , Persona de Mediana Edad , Fenotipo , ARN Mensajero/análisis , Medición de Riesgo , Índice de Severidad de la Enfermedad
19.
Oncogene ; 23(49): 8088-96, 2004 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-15361832

RESUMEN

Kruppel-like factor 2 (KLF2) is a member of the KLF family of zinc-finger transcription factors and is involved in maintaining T-cell quiescence, regulating preadipocyte differentiation, endothelial cell function and lung development. We used a tetracycline-inducible system in Jurkat T leukemia cells to study the biological role of KLF2 in cellular growth and differentiation. Our results show that expression of KLF2 inhibits cell growth in autonomously proliferating Jurkat cells. Further, 3H-thymidine uptake assays indicate that KLF2 inhibits DNA synthesis in these cells. Moreover, both activation and inhibitory domains are required for KLF2 to suppress Jurkat cell proliferation. In addition, KLF2 upregulates p21WAF1/CIP1 expression. Additionally, we found that KLF2 upregulates p21WAF1/CIP1 promoter activity in Jurkat, HepG2 and SW480 cells. Our analysis shows that the potential KLF2 responsive elements are located between -124 and -60 of the p21WAF1/CIP1 promoter. The sole CACCC site, a sequence recognized by KLF2, in this region is not the element responsive to KLF2. Finally, we determined that the Sp1-3-binding site is the functional responsive element of KLF2 in the p21WAF1/CIP1 promoter, and we conclude that KLF2 directly regulates p21WAF1/CIP1 expression.


Asunto(s)
Ciclinas/genética , Transactivadores/fisiología , División Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Proteínas de Unión al ADN/metabolismo , Regulación Leucémica de la Expresión Génica , Humanos , Células Jurkat , Factores de Transcripción de Tipo Kruppel , Regiones Promotoras Genéticas , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción Sp3 , Factores de Transcripción/metabolismo , Regulación hacia Arriba
20.
J Invest Dermatol ; 119(3): 678-83, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12230512

RESUMEN

Gene targeting of a member of small leucine-rich repeat proteoglycans demonstrates that collagen fibrillogenesis is mediated by a set of extracellular matrix components, which interact with collagen. Collagen-associated protein dermatopontin knockout mice were generated in order to analyze the biologic involvement of dermatopontin in the formation of collagen fibrils. Although dermatopontin-null mice did not exhibit any obvious anatomical abnormality, skin elasticity was increased. Skin tensile tests revealed that the initial elastic modulus was 57% lower in dermatopontin-null mice than in wild-type mice, and that maximum tensile strength was similar. Remarkably, light microscopy study showed a significant decrease in the relative thickness of the dermis in dermatopontin-null mice compared with wild-type mice (45.2 +/- 3.09% and 57.8 +/- 4.25%, respectively). The skin collagen content was 40% lower in dermatopontin-null than in wild-type mice. Collagen fibrils in dermatopontin-null mice showed a great variety in diameter and irregular contours under the electron microscope. These data indicate that dermatopontin plays a critical role in elasticity of skin and collagen accumulation attributed to collagen fibrillogenesis in vivo.


Asunto(s)
Proteínas Portadoras/genética , Moléculas de Adhesión Celular/genética , Colágeno/metabolismo , Dermis/patología , Matriz Extracelular/metabolismo , Animales , Proteoglicanos Tipo Condroitín Sulfato , Colágeno/ultraestructura , Dermis/metabolismo , Elasticidad , Matriz Extracelular/patología , Proteínas de la Matriz Extracelular , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica , Mutagénesis/fisiología , Resistencia a la Tracción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...