Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
MAbs ; 12(1): 1755000, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32329655

RESUMEN

The role of brain-derived neurotrophic factor (BDNF) signaling in chronic pain has been well documented. Given the important central role of BDNF in long term plasticity and memory, we sought to engineer a high affinity, peripherally-restricted monoclonal antibody against BDNF to modulate pain. BDNF shares 100% sequence homology across human and rodents; thus, we selected chickens as an alternative immune host for initial antibody generation. Here, we describe the affinity optimization of complementarity-determining region-grafted, chicken-derived R3bH01, an anti-BDNF antibody specifically blocking the TrkB receptor interaction. Antibody optimization led to the identification of B30, which has a > 300-fold improvement in affinity based on BIAcore, an 800-fold improvement in potency in a cell-based pERK assay and demonstrates exquisite selectivity over related neurotrophins. Affinity improvements measured in vitro translated to in vivo pharmacological activity, with B30 demonstrating a 30-fold improvement in potency over parental R3bH01 in a peripheral nerve injury model. We further demonstrate that peripheral BDNF plays a role in maintaining the plasticity of sensory neurons following nerve damage, with B30 reversing neuron hyperexcitability associated with heat and mechanical stimuli in a dose-dependent fashion. In summary, our data demonstrate that effective sequestration of BDNF via a high affinity neutralizing antibody has potential utility in modulating the pathophysiological mechanisms that drive chronic pain states.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Afinidad de Anticuerpos/inmunología , Factor Neurotrófico Derivado del Encéfalo/inmunología , Dolor Crónico/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Factor Neurotrófico Derivado del Encéfalo/antagonistas & inhibidores , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Pollos , Dolor Crónico/fisiopatología , Dolor Crónico/prevención & control , Modelos Animales de Enfermedad , Humanos , Masculino , Dimensión del Dolor , Traumatismos de los Nervios Periféricos/metabolismo , Traumatismos de los Nervios Periféricos/fisiopatología , Traumatismos de los Nervios Periféricos/prevención & control , Unión Proteica/efectos de los fármacos , Ratas Sprague-Dawley , Receptor trkB/metabolismo
2.
Oncotarget ; 8(27): 43662-43677, 2017 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-28599273

RESUMEN

Treatment of BCR-ABL+ human leukemia has been significantly improved by ABL tyrosine kinase inhibitors (TKIs), but they are not curative for most patients and relapses are frequently associated with BCR-ABL mutations, warranting new targets for improved treatments. We have now demonstrated that protein expression of human estrogen receptor alpha 36 (ERα36), an alternative splicing variant of human estrogen receptor alpha 66 (ERα66), is highly increased in TKI-insensitive CD34+ chronic myeloid leukemia (CML) cells and BCR-ABL-T315I mutant cells, and is abnormally localized in plasma membrane and cytoplasm. Interestingly, new pre-clinically-validated analogs of Icaritin (SNG162 and SNG1153), which target abnormal ERα36 activity, inhibit cell growth and induce apoptosis of BCR-ABL+ leukemic cells, particularly BCR-ABL-T315I mutant cells. A combination of SNG inhibitors and TKI selectively eliminates treatment-naïve TKI-insensitive stem/progenitor cells while sparing healthy counterparts. Oral TKI dasatinib combined with potent SNG1153 inhibitor effectively eliminates infiltrated BCR-ABL+ blast cells and enhances survival of mice. Importantly, a unique mechanism of SNG inhibition was uncovered by demonstrating a marked interruption of the BCR-ABLTyr177-GRB2 interaction, leading to inhibition of the downstream RAS/MAPK pathway. This new combination therapy may lead to more effective disease eradication, especially in patients at high risk of TKI resistance and disease progression.


Asunto(s)
Sustitución de Aminoácidos , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Proteína Adaptadora GRB2/metabolismo , Mutación , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Línea Celular Tumoral , Membrana Celular , Proliferación Celular/efectos de los fármacos , Cromanos/farmacología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Expresión Génica , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/mortalidad , Modelos Biológicos , Fosforilación , Unión Proteica , Transporte de Proteínas
3.
Oncotarget ; 7(29): 45158-45170, 2016 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-27281614

RESUMEN

Lung cancer is the leading cause of cancer-related death in both men and women. Lung cancer contains a small population of cancer cells with stem-like features known as cancer stem cells (CSCs). CSCs are often more resistant to current therapeutic treatments. Thus, it is urgent to develop a novel agent that is able to inhibit CSCs growth. In this study, we examined the ability of SNG1153, a novel chemical agent to inhibit the growth of lung CSCs. We found that SNG1153 inhibited growth and induced apoptosis in established lung cancer cells. We also found that SNG1153 inhibited the tumorsphere formation and decreased CD133-positive (lung CSC marker) cancer cells. SNG1153 was able to attenuate tumor formation in NOD/SCID (non-obese diabetic/severe combined immunodeficient) mice injected with lung tumorsphere cells. We further demonstrated that SNG1153 induced ß-catenin phosphorylation and down-regulated ß-catenin. Our results thus demonstrate that SNG1153 effectively inhibits the growth of lung CSCs and suggest that SNG1153 may be a novel therapeutic agent to treat human lung cancer.


Asunto(s)
Antineoplásicos/farmacología , Cromanos/farmacología , Epimedium/química , Neoplasias Pulmonares/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Flavonoides/farmacología , Humanos , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones SCID , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/análisis , beta Catenina/fisiología
4.
Oncotarget ; 6(31): 31927-43, 2015 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-26376676

RESUMEN

Tumor-initiating cell (TIC) is a subpopulation of cells in tumors that are responsible for tumor initiation and progression. Recent studies indicate that hepatocellular carcinoma-initiating cells (HCICs) confer the high malignancy, recurrence and multi-drug resistance in hepatocellular carcinoma (HCC). In this study, we found that Icaritin, a prenylflavonoid derivative from Epimedium Genus, inhibited malignant growth of HCICs. Icaritin decreased the proportion of EpCAM-positive (a HCICs marker) cells, suppressed tumorsphere formation in vitro and tumor formation in vivo. We also found that Icaritin reduced expression of Interleukin-6 Receptors (IL-6Rs), attenuated both constitutive and IL-6-induced phosphorylation of Janus-activated kinases 2 (Jak2) and Signal transducer and activator of transcription 3 (Stat3), and inhibited Stat3 downstream genes, such as Bmi-1 and Oct4. The inhibitory activity of Icaritin in HCICs was augmented by siRNA-mediated silencing of Stat3 but attenuated by constitutive activation of Stat3.Taken together, our results indicate that Icaritin is able to inhibit malignant growth of HCICs and suggest that Icaritin may be developed into a novel therapeutic agent for effective treatment of HCC.


Asunto(s)
Carcinoma Hepatocelular/patología , Flavonoides/farmacología , Interleucina-6/metabolismo , Janus Quinasa 2/metabolismo , Neoplasias Hepáticas/patología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Animales , Antineoplásicos/farmacología , Apoptosis , Western Blotting , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Proliferación Celular , Femenino , Humanos , Técnicas para Inmunoenzimas , Interleucina-6/genética , Janus Quinasa 2/genética , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Fosforilación , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Comp Neurol ; 523(13): 1913-24, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25753355

RESUMEN

Tumor necrosis factor receptor-associated factor 2 (TRAF2)- and noncatalytic region of tyrosine kinase (NCK)-interacting kinase (TNIK) has been identified as an interactor in the psychiatric risk factor, Disrupted in Schizophrenia 1 (DISC1). As a step toward deciphering its function in the brain, we performed high-resolution light and electron microscopic immunocytochemistry. We demonstrate here that TNIK is expressed in neurons throughout the adult mouse brain. In striatum and cerebral cortex, TNIK concentrates in dendritic spines, especially in the vicinity of the lateral edge of the synapse. Thus, TNIK is highly enriched at a microdomain critical for glutamatergic signaling.


Asunto(s)
Encéfalo/citología , Espinas Dendríticas/metabolismo , Regulación de la Expresión Génica/genética , Neuronas/citología , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Encéfalo/metabolismo , Colina O-Acetiltransferasa/metabolismo , Espinas Dendríticas/genética , Espinas Dendríticas/ultraestructura , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Inmunoelectrónica , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/ultraestructura , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Ácido gamma-Aminobutírico/metabolismo
6.
Mol Pharm ; 12(6): 1730-7, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25423493

RESUMEN

The use of predictive preclinical models in drug discovery is critical for compound selection, optimization, preclinical to clinical translation, and strategic decision-making. Trophoblast glycoprotein (TPBG), also known as 5T4, is the therapeutic target of several anticancer agents currently in clinical development, largely due to its high expression in tumors and low expression in normal adult tissues. In this study, mice were engineered to express human TPBG under endogenous regulatory sequences by replacement of the murine Tpbg coding sequence. The gene replacement was considered functional since the hTPBG knockin (hTPBG-KI) mice did not exhibit clinical observations or histopathological phenotypes that are associated with Tpbg gene deletion, except in rare instances. The expression of hTPBG in certain epithelial cell types and in different microregions of the brain and spinal cord was consistent with previously reported phenotypes and expression patterns. In pharmacokinetic studies, the exposure of a clinical-stage anti-TPBG antibody-drug conjugate (ADC), A1mcMMAF, was lower in hTPBG-KI versus wild-type animals, which was evidence of target-related increased clearance in hTPBG-KI mice. Thus, the hTPBG-KI mice constitute an improved system for pharmacology studies with current and future TPBG-targeted therapies and can generate more precise pharmacokinetic and pharmacodynamic data. In general the strategy of employing gene replacement to improve pharmacokinetic assessments should be broadly applicable to the discovery and development of ADCs and other biotherapeutics.


Asunto(s)
Inmunoconjugados/farmacocinética , Glicoproteínas de Membrana/metabolismo , Animales , Anticuerpos Monoclonales Humanizados/farmacocinética , Humanos , Inmunohistoquímica , Glicoproteínas de Membrana/genética , Ratones , Ratones Transgénicos , Fenotipo
7.
Am J Physiol Endocrinol Metab ; 306(10): E1176-87, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24714397

RESUMEN

Glycerol-3-phosphate acyltransferases (GPATs) catalyze the first step in the synthesis of glycerolipids and glycerophospholipids. Microsomal GPAT, the major GPAT activity, is encoded by at least two closely related genes, GPAT3 and GPAT4. To investigate the in vivo functions of GPAT3, we generated Gpat3-deficient (Gpat3(-/-)) mice. Total GPAT activity in white adipose tissue of Gpat3(-/-) mice was reduced by 80%, suggesting that GPAT3 is the predominant GPAT in this tissue. In liver, GPAT3 deletion had no impact on total GPAT activity but resulted in a 30% reduction in N-ethylmaleimide-sensitive GPAT activity. The Gpat3(-/-) mice were viable and fertile and exhibited no obvious metabolic abnormalities on standard laboratory chow. However, when fed a high-fat diet, female Gpat3(-/-) mice showed decreased body weight gain and adiposity and increased energy expenditure. Increased energy expenditure was also observed in male Gpat3(-/-) mice, although it was not accompanied by a significant change in body weight. GPAT3 deficiency lowered fed, but not fasted, glucose levels and tended to improve glucose tolerance in diet-induced obese male and female mice. On a high-fat diet, Gpat3(-/-) mice had enlarged livers and displayed a dysregulation in cholesterol metabolism. These data establish GPAT3 as the primary GPAT in white adipose tissue and reveal an important role of the enzyme in regulating energy, glucose, and lipid homeostasis.


Asunto(s)
Tejido Adiposo Blanco/enzimología , Colesterol/metabolismo , Metabolismo Energético/genética , Glicerol-3-Fosfato O-Aciltransferasa/metabolismo , Obesidad/enzimología , Animales , Dieta/efectos adversos , Femenino , Glicerol-3-Fosfato O-Aciltransferasa/genética , Homeostasis/genética , Metabolismo de los Lípidos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/genética
8.
PLoS One ; 8(9): e74094, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24040176

RESUMEN

HTRA1 is a member of the High Temperature Requirement (HTRA1) family of serine proteases, which play a role in several biological and pathological processes. In part, HTRA1 regulation occurs by inhibiting the TGF-ß signaling pathway, however the mechanism of inhibition has not been fully defined. Previous studies have shown that HTRA1 is expressed in a variety of tissues, including sites of skeletal development. HTRA1 has also been implicated in the process of bone formation, although the precise manner of regulation is still unknown. This study investigated how HTRA1 regulates TGF-ß signaling and examined the in vivo effects of the loss of HTRA1. We demonstrated that recombinant HTRA1 was capable of cleaving both type II and type III TGF-ß receptors (TßRII and TßRIII) in vitro in a dose-dependent manner, but it did not affect the integrity of TßRI or TGF-ß. Overexpression of HTRA1 led to decreased levels of both TßRII and III on the cell surface but had no effect on TßRI. Silencing HTRA1 expression significantly increased TGF-ß binding to the cell surface and TGF-ß responsiveness within the cell. To examine the role of HTRA1 in vivo, we generated mice with a targeted gene deletion of HTRA1. Embryonic fibroblasts isolated from these mice displayed an increase in TGF-ß-induced expression of several genes known to promote bone formation. Importantly, the loss of HTRA1 in the knockout mice resulted in a marked increase in trabecular bone mass. This study has identified a novel regulatory mechanism by which HTRA1 antagonizes TGF-ß signaling, and has shown that HTRA1 plays a key role in the regulation of bone formation.


Asunto(s)
Osteogénesis/fisiología , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Serina Endopeptidasas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Línea Celular , Fibroblastos/metabolismo , Orden Génico , Silenciador del Gen , Serina Peptidasa A1 que Requiere Temperaturas Altas , Humanos , Masculino , Ratones , Ratones Noqueados , Unión Proteica , Proteolisis , Serina Endopeptidasas/genética , Transcripción Genética
9.
Endocrinology ; 153(9): 4290-303, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22753645

RESUMEN

Coiled-coil domain containing 80 (Ccdc80) is a secreted protein highly enriched in mouse and human white adipose tissue (WAT) that plays an important role during adipocyte differentiation in vitro. To investigate the physiological function of Ccdc80 in energy and glucose homeostasis, we generated mice in which the gene encoding Ccdc80 was disrupted. Mice lacking Ccdc80 showed increased sensitivity to diet-induced hyperglycemia and glucose intolerance while displaying reduced glucose-stimulated insulin secretion in vivo. Gene expression analysis by microarray revealed that only 10 transcripts were simultaneously altered in pancreas, skeletal muscle, and WAT from Ccdc80(-/-) mice, including some components of the circadian clock. Expression of the core clock member Arntl/Bmal1 was reduced whereas that of the oscillating transcription factors Dbp and Tef was increased in all tissues examined. Furthermore, knockdown of Ccdc80 in 3T3-L1 cells led to an increase of Dbp mRNA levels during adipocyte differentiation, suggesting that Ccdc80 might be involved in the regulation of this gene in a cell-autonomous manner. Importantly, transcriptional alterations in Ccdc80(-/-) mice were associated with changes in feeding behavior, increased caloric intake, decreased energy expenditure, and obesity. Taken together, our results suggest that Ccdc80 is a novel modulator of glucose and energy homeostasis during diet-induced obesity.


Asunto(s)
Glucosa/metabolismo , Glicoproteínas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Obesidad/metabolismo , Células 3T3-L1 , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de la Matriz Extracelular , Glicoproteínas/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Obesos , Músculo Esquelético/metabolismo , Obesidad/genética , Páncreas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
J Cell Physiol ; 219(2): 430-7, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19170073

RESUMEN

Lymphatic vessels play a key role in maintaining tissue-fluid homeostasis, immune surveillance and metastasis. The hyaluronan receptor, LYVE-1, is widely used as a molecular marker for adult and embryonic lymphatic endothelium, but its physiological functions have not yet been established in vivo. In agreement with a recent report, LYVE-1(-/-) mice, which are healthy and fertile, do not display any defects related to congenital abnormalities of the lymphatic system. One hypothesis for the absence of a phenotype in LYVE-1 null mice is that other hyaluronan receptors, such as CD44, may compensate for LYVE-1. To test this hypothesis, we created LYVE-1/CD44 double knockout mice with appropriate littermate controls. Lymphatic vessel structure and function, as determined by histological methods and intravital microscopy, show that LYVE-1(-/-), CD44(-/-) and LYVE-1(-/-)/CD44(-/-) mice are indistinguishable from wild-type mice under normal conditions. Furthermore, resolution of carrageenan-induced paw edema is comparable in all genotypes. However, LYVE-1(-/-)/CD44(-/-) mice exhibit increased edema formation in a carrageenan-induced paw inflammation model compared to wild-type mice, but not to LYVE(-/-) or CD44(-/-) mice. These data suggest that LYVE-1 and CD44 are not required for the formation or function of lymphatics, but do not rule out a role for LYVE-1 in inflammation.


Asunto(s)
Glicoproteínas/metabolismo , Receptores de Hialuranos/metabolismo , Vasos Linfáticos , Ratones Noqueados , Animales , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Femenino , Genotipo , Glicoproteínas/genética , Receptores de Hialuranos/genética , Vasos Linfáticos/anatomía & histología , Vasos Linfáticos/fisiología , Masculino , Proteínas de Transporte de Membrana , Ratones , Ratones Endogámicos C57BL , Fenotipo
11.
Cancer Res ; 68(7): 2081-4, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18381412

RESUMEN

The low-fidelity polymerase eta (poleta) is required for bypass of UV-induced pyrimidine dimers inserting adenine nucleotides opposite these lesions. Mutations in the poleta gene are responsible for the genetic defect in xeroderma pigmentosum variant patients. To study if the lack of poleta significantly elevates spontaneous mutation frequency in various organs and tissues of the mouse, we crossed poleta-deficient mice with transgenic mice harboring a chromosomally integrated lacZ-plasmid reporter construct. In cultured embryonic fibroblasts from the lacZ-poleta(-/-) mice, 2.5 J/m(2) UV irradiation induced approximately 5-fold more mutations than in cells from lacZ control mice, in which an approximately 3-fold increase in mutation frequency was found compared with the normal level. Whereas untreated cells harbored mainly 1-bp deletions, UV induced both transitions and transversions, with the latter type more highly represented in the poleta-null cells than in the controls. No difference in mutation induction between the poleta-null cells and the wild-type cells was observed after treatment with N-ethyl-N-nitrosourea. Having shown the validity of the lacZ model to accurately identify poleta-associated mutagenesis, we then determined the mutant frequency at the lacZ locus in liver, spleen, and small intestine of 12-month-old animals. No differences were found between poleta-null, heterozygous, or littermate control mice. We conclude that the poleta defect is specific for UV damage and has no effect on in vivo mutagenesis in mice.


Asunto(s)
ADN Polimerasa Dirigida por ADN/deficiencia , Mutación Puntual , Animales , Etilnitrosourea , Fibroblastos/efectos de los fármacos , Fibroblastos/efectos de la radiación , Operón Lac , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Plásmidos/genética , Rayos Ultravioleta
12.
Proc Natl Acad Sci U S A ; 103(48): 18083-8, 2006 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-17114294

RESUMEN

DNA polymerase iota (pol iota) is a conserved Y family enzyme that is implicated in translesion DNA synthesis (TLS) but whose cellular functions remain uncertain. To test the hypothesis that pol iota performs TLS in cells, we compared UV-induced mutagenesis in primary fibroblasts derived from wild-type mice to mice lacking functional pol eta, pol iota, or both. A deficiency in mouse DNA polymerase eta (pol eta) enhanced UV-induced Hprt mutant frequencies. This enhanced UV-induced mutagenesis and UV-induced mutagenesis in wild-type cells were strongly diminished in cells deficient in pol iota, indicating that pol iota participates in the bypass of UV photoproducts in cells. Moreover, a clear strand bias among UV-induced base substitutions was observed in wild-type cells that was diminished in pol eta- and pol iota-deficient mouse cells and abolished in cells deficient in both enzymes. These data suggest that these enzymes bypass UV photoproducts in an asymmetric manner. To determine whether pol iota status affects cancer susceptibility, we compared the UV-induced skin cancer susceptibility of wild-type mice to mice lacking functional pol eta, pol iota, or both. Although pol iota deficiency alone had no effect, UV-induced skin tumors in pol eta-deficient mice developed 4 weeks earlier in mice concomitantly deficient in pol iota. Collectively, these data reveal functions for pol iota in bypassing UV photoproducts and in delaying the onset of UV-induced skin cancer.


Asunto(s)
Daño del ADN , ADN Polimerasa Dirigida por ADN/metabolismo , Mutagénesis/genética , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/patología , Rayos Ultravioleta , Animales , Supervivencia Celular/efectos de la radiación , Células Cultivadas , ADN Polimerasa Dirigida por ADN/deficiencia , ADN Polimerasa Dirigida por ADN/genética , Fibroblastos , Ratones , Ratones Noqueados , Sensibilidad y Especificidad , Neoplasias Cutáneas/genética , ADN Polimerasa iota
13.
Cancer Res ; 66(7): 3360-4, 2006 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-16585153

RESUMEN

Recent improvements in diagnostic methods have opened avenues for detection and treatment of (pre)malignant lesions at early stages. However, due to the lack of spontaneous tumor models that both mimic human carcinogenesis and allow direct optical imaging of the vasculature, little is known about the function of blood and lymphatic vessels during the early stages of cancer development. Here, we used a spontaneous carcinogenesis model in the skin of DNA polymerase eta-deficient mice and found that interstitial fluid pressure was already elevated in the hyperplastic/dysplastic stage. This was accompanied by angiogenic blood vasculature that exhibited altered permeability, vessel compression, and decreased alpha-smooth muscle actin-positive perivascular cell coverage. In addition, the lymphatic vessels in hyperplastic/dysplastic lesions were partly compressed and nonfunctional. These novel insights may aid early detection and treatment strategies for cancer.


Asunto(s)
Líquido Extracelular/fisiología , Vasos Linfáticos/fisiología , Lesiones Precancerosas/irrigación sanguínea , Neoplasias Cutáneas/irrigación sanguínea , Animales , Vasos Sanguíneos/fisiología , Transformación Celular Neoplásica , ADN Polimerasa Dirigida por ADN/deficiencia , Presión Hidrostática , Hiperplasia , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Lesiones Precancerosas/metabolismo , Neoplasias Cutáneas/metabolismo
14.
Cancer Res ; 66(1): 87-94, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16397220

RESUMEN

Xeroderma pigmentosum variant (XPV) patients with mutations in the DNA polymerase eta (pol eta) gene are hypersensitive to sunlight and have greatly increased susceptibility to sunlight-induced skin cancer. Consistent with the ability of Pol eta to efficiently bypass UV light-induced cyclobutane pyrimidine dimers, XPV cells lacking Pol eta have diminished capacity to replicate UV-damaged DNA and are sensitive to UV light-induced killing and mutagenesis. To better understand these and other Pol eta functions, we generated Pol eta-deficient mice. Mice homozygous for a null mutation in pol eta are viable, fertile, and do not show any obvious spontaneous defects during the first year of life. However, fibroblasts derived from these mutant mice are sensitive to killing by exposure to UV light, and all Pol eta-deficient mice develop skin tumors after UV irradiation, in contrast to the wild-type littermate controls that did not develop such tumors. These results and biochemical studies of translesion synthesis by mouse Pol eta indicate that Pol eta-dependent bypass of cyclobutane pyrimidine dimers suppresses UV light-induced skin cancer in mice. Moreover, 37.5% of pol eta heterozygous mice also developed skin cancer during 5 months after a 5-month exposure to UV light, suggesting that humans who are heterozygous for mutations in pol eta may also have an increased risk of skin cancer.


Asunto(s)
ADN Polimerasa Dirigida por ADN/deficiencia , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/etiología , Rayos Ultravioleta , Animales , Supervivencia Celular/efectos de la radiación , ADN Polimerasa Dirigida por ADN/genética , Embrión de Mamíferos , Femenino , Fertilidad/genética , Fibroblastos/citología , Fibroblastos/efectos de la radiación , Eliminación de Gen , Humanos , Ratones , Embarazo , Neoplasias Cutáneas/genética
15.
J Biol Chem ; 279(22): 23525-35, 2004 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-15039436

RESUMEN

H1 linker histones are involved in packaging chromatin into 30-nm fibers and higher order structures. Most eukaryotic cells contain nearly one H1 molecule for each nucleosome core particle. Male germ cells in mammals contain large amounts of a germ cell-specific linker histone, HIST1HT, herein denoted H1t, which is particularly abundant in pachytene spermatocytes. Despite its abundance in male germ cells and significant divergence in primary sequence from other H1 subtypes, inactivation of the H1t gene in mice showed that it is not required for spermatogenesis. Analysis of germ cell chromatin from H1t null mice showed that other H1 subtypes, especially the testis-enriched HIST1H1A, herein denoted as the H1a subtype, were able to compensate for the absence of H1t to maintain a normal total H1 to nucleosome core ratio. To disrupt the compensation, we generated H1t and H1a double null mice by two sequential gene-targeting steps in embryonic stem cells. Elimination of both H1t and H1a led to a 25% decrease in the ratio of H1 to nucleosome cores in double null germ cells. Surprisingly, the reduction in H1 did not perturb spermatogenesis or produce detectable defects in meiotic processes. Microarray analysis of gene expression showed that the reduced linker histone levels did not affect global gene expression, but it did cause changes in expression of specific genes. Our results indicate that a partial reduction in linker histone-nucleosome core particle stoichiometry is tolerated in developing male germ cells.


Asunto(s)
Histonas/fisiología , Espermatocitos/fisiología , Animales , Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Espermatogénesis/genética
16.
Proc Natl Acad Sci U S A ; 100(10): 5920-5, 2003 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-12719535

RESUMEN

Posttranslational modifications and remodeling of nucleosomes are critical factors in the regulation of transcription. Higher-order folding of chromatin also is likely to contribute to the control of gene expression, but the absence of a detailed description of the structure of the chromatin fiber has impaired progress in this area. Mammalian somatic cells contain a set of H1 linker-histone subtypes, H1 (0) and H1a to H1e, that bind to nucleosome core particles and to the linker DNA between nucleosomes. To determine whether the H1 histone subtypes play differential roles in the regulation of gene expression, we combined mice lacking specific H1 histone subtypes with mice carrying transgenes subject to position effects. Because position effects result from the unique chromatin structure created by the juxtaposition of regulatory elements in the transgene and at the site of integration, transgenes can serve as exquisitely sensitive indicators of chromatin structure. We report that some, but not all, linker histones can attenuate or accentuate position effects. The results suggest that the linker-histone subtypes play differential roles in the control of gene expression and that the sequential arrangement of the linker histones on the chromatin fiber might regulate higher-order chromatin structure and fine-tune expression levels.


Asunto(s)
Anemia Hemolítica/sangre , Globinas/genética , Globinas/metabolismo , Histonas/genética , Anemia Hemolítica/inducido químicamente , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/fisiología , Eliminación de Gen , Regulación de la Expresión Génica , Histonas/metabolismo , Hibridación Fluorescente in Situ , Ratones , Ratones Transgénicos , Fenilhidrazinas , Procesamiento Proteico-Postraduccional , Bazo/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA