Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
BMC Cancer ; 17(1): 203, 2017 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-28320353

RESUMEN

BACKGROUND: About 75-80% of breast tumors express the estrogen receptor alpha (ER-α) and are treated with endocrine-target therapeutics, making this the premier therapeutic modality in the breast cancer clinic. However, acquired resistance is common and about 20% of resistant tumors loose ER-α expression via unknown mechanisms. Inhibition of ER-α loss could improve endocrine therapeutic efficacy, benefiting a significant number of patients. Here we test whether tumor hypoxia might commonly produce ER-α loss. METHODS: Using standard molecular and cellular biological assays and a work station/incubator with controllable oxygen levels, we analyze the effects of hypoxia on ER-α protein, mRNA, and transcriptional activity in a panel of independently-derived ER-α positive cell lines. These lines were chosen to represent the diverse genetic backgrounds and mutations commonly present in ER-α positive tumors. Using shRNA-mediated knockdown and overexpression studies we also elucidate the role of hypoxia-inducible factor 1-alpha (HIF-1α) in the hypoxia-induced decrease in ER-α abundance. RESULTS: We present the first comprehensive overview of the effects of bona fide low environmental oxygen (hypoxia) and HIF-1α activity on ER-α abundance and transcriptional activity. We find that stabilized HIF-1α induces rapid loss of ER-α protein in all members of our diverse panel of breast cancer cell lines, which involves proteolysis rather than transcriptional repression. Reduced ER-α severely attenuates ER-α directed transcription, and inhibits cell proliferation without overt signs of cell death in the cell lines tested, despite their varying genomic backgrounds. CONCLUSIONS: These studies reveal a common hypoxia response that produces reduced ER-α expression and cell cycle stalling, and demonstrate a common role for HIF-1α in ER-α loss. We hypothesize that inhibitors of HIF-1α or the proteasome might stabilize ER-α expression in breast tumors in vivo, and work in combination with endocrine therapies to reduce resistance. Our data also suggests that disease re-occurrence in patients with ER-α positive tumors may arise from tumor cells chronically resident in hypoxic environments. We hypothesize that these non-proliferating cells may survive undetected until conditions change to oxygenate the environment, or cells eventually switch to proliferation via other signaling pathways.


Asunto(s)
Neoplasias de la Mama/genética , Receptor alfa de Estrógeno/metabolismo , Regulación Neoplásica de la Expresión Génica , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Hipoxia de la Célula , Línea Celular Tumoral , Proliferación Celular , Receptor alfa de Estrógeno/genética , Femenino , Expresión Génica , Silenciador del Gen , Genoma Humano , Humanos , Recurrencia Local de Neoplasia/metabolismo
2.
Bioorg Med Chem ; 22(2): 711-20, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24387866

RESUMEN

Two novel scaffolds, 4-pyridylanilinothiazoles (PAT) and 3-pyridylphenylsulfonyl benzamides (PPB), previously identified as selective cytotoxins for von Hippel-Lindau-deficient Renal Carcinoma cells, were used as templates to prepare affinity chromatography reagents to aid the identification of the molecular targets of these two classes. Structure-activity data and computational models were used to predict possible points of attachment for linker chains. In the PAT class, Click coupling of long chain azides with 2- and 3-pyridylanilinothiazoleacetylenes gave triazole-linked pyridylanilinothiazoles which did not retain the VHL-dependent selectivity of parent analogues. For the PPB class, Sonagashira coupling of 4-iodo-(3-pyridylphenylsulfonyl)benzamide with a propargyl hexaethylene glycol carbamate gave an acetylene which was reduced to the corresponding alkyl 3-pyridylphenylsulfonylbenzamide. This reagent retained the VHL-dependent selectivity of the parent analogues and was successfully utilized as an affinity reagent.


Asunto(s)
Antineoplásicos/farmacología , Benzamidas/farmacología , Carcinoma de Células Renales/tratamiento farmacológico , Diseño de Fármacos , Neoplasias Renales/tratamiento farmacológico , Piridinas/farmacología , Sulfonas/farmacología , Tiazoles/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Benzamidas/síntesis química , Benzamidas/química , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cromatografía de Afinidad , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Neoplasias Renales/patología , Modelos Moleculares , Estructura Molecular , Piridinas/síntesis química , Piridinas/química , Relación Estructura-Actividad , Sulfonas/síntesis química , Sulfonas/química , Tiazoles/síntesis química , Tiazoles/química
4.
J Biol Chem ; 285(35): 26852-26860, 2010 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-20581113

RESUMEN

Toxoplasma gondii is an intracellular protozoan parasite that can cause devastating disease in fetuses and immune-compromised individuals. We previously reported that the alpha subunit of the host cell transcription factor, hypoxia-inducible factor-1 (HIF-1), is up-regulated by infection and necessary for Toxoplasma growth. Under basal conditions, HIF-1alpha is constitutively expressed but rapidly targeted for proteasomal degradation after two proline residues are hydroxylated by a family of prolyl hydroxylases (PHDs). The PHDs are alpha-ketoglutarate-dependent dioxygenases that have low K(m) values for oxygen, making them important cellular oxygen sensors. Thus, when oxygen levels decrease, HIF-1alpha is not hydroxylated, and HIF-1 is activated. How Toxoplasma activates HIF-1 under normoxic conditions remains unknown. Here, we report that Toxoplasma infection increases HIF-1alpha stability by preventing HIF-1alpha prolyl hydroxylation. Infection significantly decreases PHD2 abundance, which is the key prolyl hydroxylase for regulating HIF-1alpha. The effects of Toxoplasma on HIF-1alpha abundance and prolyl hydroxylase activity require activin-like receptor kinase signaling. Finally, parasite growth is severely diminished when signaling from this family of receptors is inhibited. Together, these data indicate that PHD2 is a key host cell factor for T. gondii growth and represent a novel mechanism by which a microbial pathogen subverts host cell signaling and transcription to establish its replicative niche.


Asunto(s)
Receptores de Activinas Tipo I/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Procolágeno-Prolina Dioxigenasa/metabolismo , Toxoplasma/metabolismo , Toxoplasmosis/mortalidad , Animales , Células HeLa , Humanos , Hidroxilación , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Oxígeno/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica
5.
Blood ; 113(22): 5568-74, 2009 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-19336759

RESUMEN

The molecular mechanism of autocrine regulation of vascular endothelial growth factor (VEGF) in chronic lymphocytic leukemia (CLL) B cells is unknown. Here, we report that CLL B cells express constitutive levels of HIF-1alpha under normoxia. We have examined the status of the von Hippel-Lindau gene product (pVHL) that is responsible for HIF-1alpha degradation and found it to be at a notably low level in CLL B cells compared with normal B cells. We demonstrate that the microRNA, miR-92-1, overexpressed in CLL B cells, can target the VHL transcript to repress its expression. We found that the stabilized HIF-1alpha can form an active complex with the transcriptional coactivator p300 and phosphorylated-STAT3 at the VEGF promoter and recruit RNA polymerase II. This is initial evidence that pVHL, without any genetic alteration, can be regulated by microRNA and explains the aberrant autocrine VEGF secretion in CLL.


Asunto(s)
Factor 1 Inducible por Hipoxia/genética , Leucemia Linfocítica Crónica de Células B/genética , MicroARNs/fisiología , Factor A de Crecimiento Endotelial Vascular/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Linfocitos B/metabolismo , Linfocitos B/patología , Técnicas de Cultivo de Célula , Núcleo Celular/metabolismo , Células Cultivadas , Regulación Leucémica de la Expresión Génica , Humanos , Hidroxilación/genética , Factor 1 Inducible por Hipoxia/metabolismo , Factor 1 Inducible por Hipoxia/fisiología , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Oxigenasas de Función Mixta/metabolismo , Unión Proteica , Procesamiento Proteico-Postraduccional/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/fisiología , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Factores de Transcripción p300-CBP/metabolismo
6.
Bioorg Med Chem ; 19(11): 3347-56, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21561782

RESUMEN

We recently identified a class of pyridyl aniline thiazoles (PAT) that displayed selective cytotoxicity for von Hippel-Lindau (VHL) deficient renal cell carcinoma (RCC) cells in vitro and in vivo. Structure-activity relationship (SAR) studies were used to develop a comparative molecular field analysis (CoMFA) model that related VHL-selective potency to the three-dimensional arrangement of chemical features of the chemotype. We now report the further molecular alignment-guided exploration of the chemotype to discover potent and selective PAT analogues. The contribution of the central thiazole ring was explored using a series of five- and six-membered ring heterocyclic replacements to vary the electronic and steric interactions in the central unit. We also explored a positive steric CoMFA contour adjacent to the pyridyl ring using Pd-catalysed cross-coupling Suzuki-Miyaura, Sonogashira and nucleophilic displacement reactions to prepare of a series of aryl-, alkynyl-, alkoxy- and alkylamino-substituted pyridines, respectively. In vitro potency and selectivity were determined using paired RCC cell lines: the VHL-null cell line RCC4 and the VHL-positive cell line RCC4-VHL. Active analogues selectively induced autophagy in RCC4 cells. We have used the new SAR data to further develop the CoMFA model, and compared this to a 2D-QSAR method. Our progress towards realising the therapeutic potential of this chemotype as a targeted cytotoxic therapy for the treatment of RCC by exploiting the absence of the VHL tumour suppressor gene is reported.


Asunto(s)
Compuestos de Anilina/química , Autofagia , Carcinoma de Células Renales/tratamiento farmacológico , Neoplasias Renales/tratamiento farmacológico , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Compuestos de Anilina/síntesis química , Compuestos de Anilina/toxicidad , Humanos , Modelos Moleculares , Piridinas/química , Relación Estructura-Actividad Cuantitativa , Tiazoles/química , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
7.
FASEB J ; 23(3): 906-15, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19001054

RESUMEN

Classic tissue engineering paradigms are limited by the incorporation of a functional vasculature and a reliable means for reimplantation into the host circulation. We have developed a novel approach to overcome these obstacles using autologous explanted microcirculatory beds (EMBs) as bioscaffolds for engineering complex three-dimensional constructs. In this study, EMBs consisting of an afferent artery, capillary beds, efferent vein, and surrounding parenchymal tissue are explanted and maintained for 24 h ex vivo in a bioreactor that preserves EMB viability and function. Given the rapidly advancing field of stem cell biology, EMBs were subsequently seeded with three distinct stem cell populations, multipotent adult progenitor cells (MAPCs), and bone marrow and adipose tissue-derived mesenchymal stem cells (MSCs). We demonstrate MAPCs, as well as MSCs, are able to egress from the microcirculation into the parenchymal space, forming proliferative clusters. Likewise, human adipose tissue-derived MSCs were also found to egress from the vasculature and seed into the EMBs, suggesting feasibility of this technology for clinical applications. We further demonstrate that MSCs can be transfected to express a luciferase protein and continue to remain viable and maintain luciferase expression in vivo. By using the vascular network of EMBs, EMBs can be perfused ex vivo and seeded with stem cells, which can potentially be directed to differentiate into neo-organs or transfected to replace failing organs and deficient proteins.


Asunto(s)
Microcirculación/fisiología , Técnicas de Cultivo de Tejidos/métodos , Ingeniería de Tejidos/métodos , Animales , Reactores Biológicos , Femenino , Regulación Enzimológica de la Expresión Génica , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Masculino , Células Madre Mesenquimatosas/fisiología , Ratas , Ratas Desnudas , Piel/irrigación sanguínea , Transfección
8.
Mol Cell Biol ; 27(3): 912-25, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17101781

RESUMEN

Prolyl hydroxylation of hypoxible-inducible factor alpha (HIF-alpha) proteins is essential for their recognition by pVHL containing ubiquitin ligase complexes and subsequent degradation in oxygen (O(2))-replete cells. Therefore, HIF prolyl hydroxylase (PHD) enzymatic activity is critical for the regulation of cellular responses to O(2) deprivation (hypoxia). Using a fusion protein containing the human HIF-1alpha O(2)-dependent degradation domain (ODD), we monitored PHD activity both in vivo and in cell-free systems. This novel assay allows the simultaneous detection of both hydroxylated and nonhydroxylated PHD substrates in cells and during in vitro reactions. Importantly, the ODD fusion protein is regulated with kinetics identical to endogenous HIF-1alpha during cellular hypoxia and reoxygenation. Using in vitro assays, we demonstrated that the levels of iron (Fe), ascorbate, and various tricarboxylic acid (TCA) cycle intermediates affect PHD activity. The intracellular levels of these factors also modulate PHD function and HIF-1alpha accumulation in vivo. Furthermore, cells treated with mitochondrial inhibitors, such as rotenone and myxothiazol, provided direct evidence that PHDs remain active in hypoxic cells lacking functional mitochondria. Our results suggest that multiple mitochondrial products, including TCA cycle intermediates and reactive oxygen species, can coordinate PHD activity, HIF stabilization, and cellular responses to O(2) depletion.


Asunto(s)
Metabolismo Energético , Procolágeno-Prolina Dioxigenasa/metabolismo , Animales , Carcinoma de Células Renales/patología , Hipoxia de la Célula/efectos de los fármacos , Transporte de Electrón/efectos de los fármacos , Electroforesis en Gel de Poliacrilamida , Metabolismo Energético/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/farmacología , Hidroxilación/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Ratones , Mitocondrias/efectos de los fármacos , Imitación Molecular/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Prolina/química , Transporte de Proteínas/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Termodinámica , Desacopladores/farmacología , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
9.
Circulation ; 118(14 Suppl): S226-33, 2008 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-18824759

RESUMEN

BACKGROUND: During hypoxia, upregulation of hypoxia inducible factor-1 alpha transcriptional factor can activate several downstream angiogenic genes. However, hypoxia inducible factor-1 alpha is naturally degraded by prolyl hydroxylase-2 (PHD2) protein. Here we hypothesize that short hairpin RNA (shRNA) interference therapy targeting PHD2 can be used for treatment of myocardial ischemia and this process can be followed noninvasively by molecular imaging. METHODS AND RESULTS: PHD2 was cloned from mouse embryonic stem cells by comparing the homolog gene in human and rat. The best candidate shRNA sequence for inhibiting PHD2 was inserted into the pSuper vector driven by the H1 promoter followed by a separate hypoxia response element-incorporated promoter driving a firefly luciferase reporter gene. This construct was used to transfect mouse C2C12 myoblast cell line for in vitro confirmation. Compared with the control short hairpin scramble (shScramble) as control, inhibition of PHD2 increased levels of hypoxia inducible factor-1 alpha protein and several downstream angiogenic genes by >30% (P<0.01). Afterward, shRNA targeting PHD2 (shPHD2) plasmid was injected intramyocardially following ligation of left anterior descending artery in mice. Animals were randomized into shPHD2 experimental group (n=25) versus shScramble control group (n=20). Bioluminescence imaging detected plasmid-mediated transgene expression for 4 to 5 weeks. Echocardiography showed the shPHD2 group had improved fractional shortening compared with the shScramble group at Week 4 (33.7%+/-1.9% versus 28.4%+/-2.8%; P<0.05). Postmortem analysis showed increased presence of small capillaries and venules in the infarcted zones by CD31 staining. Finally, Western blot analysis of explanted hearts also confirmed that animals treated with shPHD2 had significantly higher levels of hypoxia inducible factor-1 alpha protein. CONCLUSIONS: This is the first study to image the biological role of shRNA therapy for improving cardiac function. Inhibition of PHD2 by shRNA led to significant improvement in angiogenesis and contractility by in vitro and in vivo experiments. With further validation, the combination of shRNA therapy and molecular imaging can be used to track novel cardiovascular gene therapy applications in the future.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas Inmediatas-Precoces/genética , Isquemia Miocárdica/fisiopatología , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Animales , Hipoxia de la Célula , Línea Celular , Ecocardiografía , Femenino , Factor 1 Inducible por Hipoxia/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Mediciones Luminiscentes , Ratones , Ratones Endogámicos , Mioblastos/citología , Mioblastos/metabolismo , Isquemia Miocárdica/diagnóstico por imagen , Isquemia Miocárdica/genética , Miocardio/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Plásmidos/farmacología , Procolágeno-Prolina Dioxigenasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Volumen Sistólico/efectos de los fármacos , Transfección , Regulación hacia Arriba , Función Ventricular Izquierda
10.
Cancer Res ; 67(12): 5896-905, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17575159

RESUMEN

Late-stage clear cell renal carcinoma poses a formidable clinical challenge due to the high mortality rate associated with this disease. Molecular and genetic studies have identified functional loss of the von Hippel-Lindau (VHL) gene as a frequent and crucial event in the development of the malignant phenotype of clear cell renal carcinomas. Loss of VHL function thus represents a pathognomonic molecular defect for therapeutic exploitation. The objective of this study was to evaluate the possibility of targeting VHL loss through pharmacologic means. Chromomycin A3 (ChA3) was identified through in silico analysis of existing publicly available drug profiles from the National Cancer Institute as an agent that seemed to selectively target VHL-deficient clear cell renal carcinoma cells. Genotype-selective toxicity was first determined through short-term viability assays and then confirmed with clonogenic studies. Coculture of fluorescently labeled VHL-deficient and VHL-positive cells showed discriminate killing of the VHL-deficient cells with ChA3. Mechanistically, overexpression of hypoxia-inducible factor (HIF)-2alpha in VHL-positive clear cell renal carcinoma cells phenocopied loss of VHL with respect to ChA3 toxicity, establishing ChA3 as a HIF-dependent cytotoxin. This study shows the feasibility of selectively targeting the loss of the VHL tumor suppressor gene in clear cell renal carcinoma for potential clinical benefit and may have greater ramifications in the development of new targeted therapies for the treatment of cancer and other genetic diseases.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Carcinoma de Células Renales/genética , Cromomicina A3/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Neoplasias Renales/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Algoritmos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Western Blotting , Carcinoma de Células Renales/tratamiento farmacológico , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Neoplasias Renales/tratamiento farmacológico
11.
Mol Pharmacol ; 74(1): 282-8, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18424552

RESUMEN

Hypoxia-inducible factors (HIFs) are unstable heterodimeric transcription factors and decisive elements for the transcriptional regulation of genes important in the adaptation to low-oxygen conditions. Hypoxia is the ubiquitous inducer of HIFs, stabilizing the alpha-subunit and permitting the formation of a functional HIF complex. Here, we identify (2R)-[(4-biphenylylsulfonyl)amino]-N-hydroxy-3-phenylpropionamide (BiPS), a commercially available metalloprotease-2 and -9 inhibitor, as a rapid and potent inducer of HIFs. We show that in different cell lines, BiPS induces the HIF-alpha subunit by inhibiting its degradation through stabilization of its labile oxygen-dependent degradation domain. This is achieved through the inhibition of HIF-1alpha hydroxylation. The HIF-1 complex, formed after BiPS treatment, is capable of DNA binding and activation of HIF target genes, including the expression of vascular endothelial growth factor. Because novel HIF activators have generated considerable interest in the possible treatment of different ischemic diseases, we believe that BiPS and derivative molecules could have strong therapeutic potential.


Asunto(s)
Ácidos Hidroxámicos/química , Factor 1 Inducible por Hipoxia/biosíntesis , Factor 1 Inducible por Hipoxia/metabolismo , Metaloendopeptidasas/antagonistas & inhibidores , Animales , Aorta Torácica/citología , Bovinos , Hipoxia de la Célula , Células Cultivadas , Relación Dosis-Respuesta a Droga , Genes Reporteros , Células HeLa , Humanos , Factor 1 Inducible por Hipoxia/genética , Cinética , Luciferasas de Luciérnaga/metabolismo , Masculino , Metaloproteinasa 2 de la Matriz , Metaloproteinasa 9 de la Matriz , Músculo Liso Vascular/citología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Wistar , Transfección
12.
Circulation ; 116(24): 2818-29, 2007 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-18040029

RESUMEN

BACKGROUND: Advanced age is known to impair neovascularization. Because endothelial progenitor cells (EPCs) participate in this process, we examined the effects of aging on EPC recruitment and vascular incorporation. METHODS AND RESULTS: Murine neovascularization was examined by use of an ischemic flap model, which demonstrated aged mice (19 to 24 months) had decreased EPC mobilization (percent mobilized 1.4+/-0.2% versus 0.4+/-0.1%, P<0.005) that resulted in impaired gross tissue survival compared with young mice (2 to 6 months). This decrease correlated with diminished tissue perfusion (P<0.005) and decreased CD31+ vascular density (P<0.005). Gender-mismatched bone marrow transplantation demonstrated significantly fewer chimeric vessels in aged mice (P<0.05), which confirmed a deficit in bone marrow-mediated vasculogenesis. Age had no effect on total EPC number in mice or humans. Reciprocal bone marrow transplantations confirmed that impaired neovascularization resulted from defects in the response of aged tissue to hypoxia and not from intrinsic defects in EPC function. We demonstrate that aging decreased hypoxia-inducible factor 1alpha stabilization in ischemic tissues because of increased prolyl hydroxylase-mediated hydroxylation (P<0.05) and proteasomal degradation. This resulted in a diminished hypoxia response, including decreased stromal cell-derived factor 1 (P<0.005) and vascular endothelial growth factor (P<0.0004). This effect can be reversed with the iron chelator deferoxamine, which results in hypoxia-inducible factor 1alpha stabilization and increased tissue survival. CONCLUSIONS: Aging impairs EPC trafficking to sites of ischemia through a failure of aged tissues to normally activate the hypoxia-inducible factor 1alpha-mediated hypoxia response.


Asunto(s)
Trasplante de Médula Ósea , Endotelio Vascular/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Trasplante de Células Madre , Células Madre/fisiología , Envejecimiento/fisiología , Animales , Endotelio Vascular/crecimiento & desarrollo , Movilización de Célula Madre Hematopoyética , Ratones , Modelos Animales , Isquemia Miocárdica/cirugía , Trasplante de Piel , Células Madre/citología , Colgajos Quirúrgicos
13.
Mol Cell Biol ; 25(15): 6415-26, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16024780

RESUMEN

Oxygen-dependent proteolysis is the primary means of regulating the hypoxia-inducible factor (HIF) family of transcription factors. The alpha-subunit of HIF factor 1 (HIF-1) contains two highly conserved oxygen-dependent degradation domains (402 ODD and 564 ODD), each of which includes a proline that is hydroxylated in the presence of oxygen, allowing the von Hippel-Lindau (VHL) E3 ubiquitin ligase to interact and target HIF-1alpha to the proteasome for degradation. Mutation of either proline is sufficient to partially stabilize HIF-1alpha under conditions of normoxia, but the specific contributions of each hydroxylation event to the regulation of HIF-1alpha are unknown. Here we show that the two ODDs of HIF-1alpha have independent yet interactive roles in the regulation of HIF-1alpha protein turnover, with the relative involvement of each ODD depending on the levels of oxygen. Using hydroxylation-specific antibodies, we found that under conditions of normoxia proline 564 is hydroxylated prior to proline 402, and mutation of proline 564 results in a significant reduction in the hydroxylation of proline 402. Mutation of proline 402, however, has little effect on the hydroxylation of proline 564. To determine whether the more rapid hydroxylation of the proline 564 under conditions of normoxia is due to a preference for the particular sequence surrounding proline 564 or for that site within the protein, we exchanged the degradation domains within the full-length HIF-1alpha protein. In these domain-swapping experiments, prolyl hydroxylase domain 1 (PHD1) and PHD2 preferentially hydroxylated the proline located in the site of the original 564 ODD, while PHD3 preferred the proline 564 sequence, regardless of its location. At limiting oxygen tensions, we found that proline 402 exhibits an oxygen-dependent decrease in hydroxylation at higher oxygen tensions relative to proline 564 hydroxylation. These results indicate that hydroxylation of proline 402 is highly responsive to physiologic changes in oxygen and, therefore, plays a more important role in HIF-1alpha regulation under conditions of hypoxia than under conditions of normoxia. Together, these findings demonstrate that each hydroxylated proline of HIF-1alpha has a distinct activity in controlling HIF-1alpha stability in response to different levels of oxygenation.


Asunto(s)
Oxígeno/fisiología , Factores de Transcripción/metabolismo , Animales , Bovinos , Línea Celular , Humanos , Hidroxilación , Subunidad alfa del Factor 1 Inducible por Hipoxia , Cinética , Ratones , Ratones Noqueados , Oxigenasas de Función Mixta/metabolismo , Mutación , Estrés Oxidativo/fisiología , Prolina/genética , Prolina/metabolismo , Estructura Terciaria de Proteína , Factores de Transcripción/química , Factores de Transcripción/genética
14.
Plast Reconstr Surg ; 141(1): 55e-67e, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29280872

RESUMEN

BACKGROUND: Cell therapy with mesenchymal stromal cells is a promising strategy for tissue repair. Restoration of blood flow to ischemic tissues is a key step in wound repair, and mesenchymal stromal cells have been shown to be proangiogenic. Angiogenesis is critically regulated by the hypoxia-inducible factor (HIF) superfamily, consisting of transcription factors targeted for degradation by prolyl hydroxylase domain (PHD)-2. The aim of this study was to enhance the proangiogenic capability of mesenchymal stromal cells and to use these modified cells to promote wound healing. METHODS: Mesenchymal stromal cells harvested from mouse bone marrow were transduced with short hairpin RNA (shRNA) against PHD-2; control cells were transduced with scrambled shRNA (shScramble) construct. Gene expression quantification, human umbilical vein endothelial cell tube formation assays, and wound healing assays were used to assess the effect of PHD knockdown mesenchymal stromal cells on wound healing dynamics. RESULTS: PHD-2 knockdown mesenchymal stromal cells overexpressed HIF-1α and multiple angiogenic factors compared to control (p < 0.05). Human umbilical vein endothelial cells treated with conditioned medium from PHD-2 knockdown mesenchymal stromal cells exhibited increased formation of capillary-like structures and enhanced migration compared with human umbilical vein endothelial cells treated with conditioned medium from shScramble-transduced mesenchymal stromal cells (p < 0.05). Wounds treated with PHD-2 knockdown mesenchymal stromal cells healed at a significantly accelerated rate compared with wounds treated with shScramble mesenchymal stromal cells (p < 0.05). Histologic studies revealed increased blood vessel density and increased cellularity in the wounds treated with PHD-2 knockdown mesenchymal stromal cells (p < 0.05). CONCLUSIONS: Silencing PHD-2 in mesenchymal stromal cells augments their proangiogenic potential in wound healing therapy. This effect appears to be mediated by overexpression of HIF family transcription factors and up-regulation of multiple downstream angiogenic factors.


Asunto(s)
Silenciador del Gen , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Neovascularización Fisiológica/fisiología , Cicatrización de Heridas/fisiología , Animales , Biomarcadores/metabolismo , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Técnicas de Silenciamiento del Gen , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba
15.
Methods Enzymol ; 435: 323-45, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17998061

RESUMEN

Tumor hypoxia is a feature common to almost all solid tumors due to malformed vasculature and inadequate perfusion. Tumor cells have evolved mechanisms that allow them to respond and adapt to a hypoxic microenvironment. The hypoxia-inducible transcription factor (HIF) family is comprised of oxygen-sensitive alpha (alpha) subunits that respond rapidly to decreased oxygen levels and oxygen-insensitive beta (beta) subunits. HIF binds to specific recognition sequences in the genome and increases the transcription of genes involved in a variety of metabolic and enzymatic pathways that are necessary for cells to respond to an oxygen-poor environment. The critical role of this family of transcriptional regulators in maintaining oxygen homeostasis is supported by multiple regulatory mechanisms that allow the cell to control the levels of HIF as well as its transcriptional activity. This review will focus on how the transcriptional activity of HIF is studied and how it can be exploited for cancer therapy.


Asunto(s)
Técnicas Genéticas , Factor 1 Inducible por Hipoxia/genética , Neoplasias/terapia , Animales , Expresión Génica , Humanos , Factor 1 Inducible por Hipoxia/análisis , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Ratones , Ratones Noqueados , Neoplasias/genética
16.
Cell Cycle ; 3(2): 160-3, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14712081

RESUMEN

Elimination or reduction of tumor burden is the primary goal of cancer therapy. Strategies to achieve this goal with the fewest adverse effects to the patient are an area of intense investigation. Elevated protein levels of hypoxia-inducible factor (HIF) are commonly found in solid tumors, while rarely found in healthy tissue. Numerous studies have suggested that HIF activity is essential for the development of solid tumors. Thus, inhibition of HIF represents an attractive therapeutic target for eradicating tumors. The search for small molecules that target and inhibit HIF activity is currently underway. We propose an alternate approach: to directly target and kill HIF-activated tumor cells. This approach is advantageous in that cells with activated HIF will be eliminated directly. Specific elimination of HIF-activated cells represents a potential mechanism for inhibiting tumor growth, with the potential advantage of sparing the patient of the normal tissue toxicity associated with current treatment options.


Asunto(s)
Muerte Celular/fisiología , Transformación Celular Neoplásica/metabolismo , Citotoxinas/metabolismo , Proteínas de Unión al ADN , Receptores de Hidrocarburo de Aril/metabolismo , Factores de Transcripción/metabolismo , Animales , Antineoplásicos/farmacología , Translocador Nuclear del Receptor de Aril Hidrocarburo , División Celular/fisiología , Hipoxia de la Célula/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia , Ratones , Modelos Moleculares , Neovascularización Patológica/metabolismo , Proteínas Nucleares/metabolismo , Oxígeno/metabolismo , Sirolimus/farmacología , Transactivadores/metabolismo , Trasplante Heterólogo , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau
17.
Nat Commun ; 6: 5990, 2015 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-25606712

RESUMEN

The exquisite sensitivity of mitotic cancer cells to ionizing radiation (IR) underlies an important rationale for the widely used fractionated radiation therapy. However, the mechanism for this cell cycle-dependent vulnerability is unknown. Here we show that treatment with IR leads to mitotic chromosome segregation errors in vivo and long-lasting aneuploidy in tumour-derived cell lines. These mitotic errors generate an abundance of micronuclei that predispose chromosomes to subsequent catastrophic pulverization thereby independently amplifying radiation-induced genome damage. Experimentally suppressing whole-chromosome missegregation reduces downstream chromosomal defects and significantly increases the viability of irradiated mitotic cells. Further, orthotopically transplanted human glioblastoma tumours in which chromosome missegregation rates have been reduced are rendered markedly more resistant to IR, exhibiting diminished markers of cell death in response to treatment. This work identifies a novel mitotic pathway for radiation-induced genome damage, which occurs outside of the primary nucleus and augments chromosomal breaks. This relationship between radiation treatment and whole-chromosome missegregation can be exploited to modulate therapeutic response in a clinically relevant manner.


Asunto(s)
Neoplasias Encefálicas/genética , Inestabilidad Cromosómica , Glioblastoma/genética , Neoplasias/radioterapia , Aneuploidia , Animales , Neoplasias Encefálicas/radioterapia , Ciclo Celular , Muerte Celular , Línea Celular Tumoral , Supervivencia Celular , Rotura Cromosómica , Segregación Cromosómica , Glioblastoma/radioterapia , Células HCT116 , Humanos , Masculino , Ratones , Ratones Desnudos , Pruebas de Micronúcleos , Mitosis/genética , Trasplante de Neoplasias , Radiación Ionizante
18.
PLoS One ; 9(4): e93373, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24695462

RESUMEN

INTRODUCTION: Hypoxia-inducible factor (HIF)-1α, part of the heterodimeric transcription factor that mediates the cellular response to hypoxia, is critical for the expression of multiple angiogenic growth factors, cell motility, and the recruitment of endothelial progenitor cells. Inhibition of the oxygen-dependent negative regulator of HIF-1α, prolyl hydroxylase domain-2 (PHD-2), leads to increased HIF-1α and mimics various cellular and physiological responses to hypoxia. The roles of PHD-2 in the epidermis and dermis have not been clearly defined in wound healing. METHODS: Epidermal and dermal specific PHD-2 knockout (KO) mice were developed in a C57BL/6J (wild type) background by crossing homozygous floxed PHD-2 mice with heterozygous K14-Cre mice and heterozygous Col1A2-Cre-ER mice to get homozygous floxed PHD-2/heterozygous K14-Cre and homozygous floxed PHD-2/heterozygous floxed Col1A2-Cre-ER mice, respectively. Ten to twelve-week-old PHD-2 KO and wild type (WT) mice were subjected to wounding and ischemic pedicle flap model. The amount of healing was grossly quantified with ImageJ software. Western blot and qRT-PCR was run on protein and RNA from primary cells cultured in vitro. RESULTS: qRT-PCR demonstrated a significant decrease of PHD-2 in keratinocytes and fibroblasts derived from tissue specific KO mice relative to control mice (*p<0.05). Western blot analysis showed a significant increase in HIF-1α and VEGF protein levels in PHD-2 KO mice relative to control mice (*p<0.05). PHD-2 KO mice showed significantly accelerated wound closure relative to WT (*p<0.05). When ischemia was analyzed at day nine post-surgery in a flap model, the PHD-2 tissue specific knockout mice showed significantly more viable flaps than WT (*p<0.05). CONCLUSIONS: PHD-2 plays a significant role in the rates of wound healing and response to ischemic insult in mice. Further exploration shows PHD-2 KO increases cellular levels of HIF-1α and this increase leads to the transcription of downstream angiogenic factors such as VEGF.


Asunto(s)
Dermis/metabolismo , Dermis/fisiopatología , Epidermis/metabolismo , Epidermis/fisiopatología , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Isquemia/fisiopatología , Cicatrización de Heridas/fisiología , Animales , Femenino , Fibroblastos/metabolismo , Fibroblastos/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Isquemia/metabolismo , Queratinocitos/metabolismo , Queratinocitos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
19.
Cancer Cell ; 24(4): 450-65, 2013 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-24094812

RESUMEN

A handful of tumor-derived cell lines form the mainstay of cancer therapeutic development, yielding drugs with an impact typically measured as months to disease progression. To develop more effective breast cancer therapeutics and more readily understand their clinical impact, we constructed a functional metabolic portrait of 46 independently derived breast cell lines. Our analysis of glutamine uptake and dependence identified a subset of triple-negative samples that are glutamine auxotrophs. Ambient glutamine indirectly supports environmental cystine acquisition via the xCT antiporter, which is expressed on one-third of triple-negative tumors in vivo. xCT inhibition with the clinically approved anti-inflammatory sulfasalazine decreases tumor growth, revealing a therapeutic target in breast tumors of poorest prognosis and a lead compound for rapid, effective drug development.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/metabolismo , Regulación Neoplásica de la Expresión Génica , Glutamina/farmacología , Neoplasias de la Mama Triple Negativas/metabolismo , Animales , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Glutamina/metabolismo , Glutatión/metabolismo , Humanos , Ratones , Pronóstico , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno
20.
Radiother Oncol ; 103(3): 388-93, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22551566

RESUMEN

BACKGROUND AND PURPOSE: For patients diagnosed with advanced renal cell carcinoma (RCC), there are few therapeutic options. Radiation therapy is predominantly used to treat metastasis and has not proven effective in the adjuvant setting for renal cancer. Furthermore, RCC is resistant to standard cytotoxic chemotherapies. Targeted anti-angiogenics are the standard of care for RCC but are not curative. Newer agents, such as mTOR inhibitors and others that induce autophagy, have shown great promise for treating RCC. Here, we investigate the potential use of the small molecule STF-62247 to modulate radiation. MATERIALS AND METHODS: Using RCC cell lines, we evaluate sensitivity to radiation in addition to agents that induce autophagic cell death by clonogenic survival assays. Furthermore, these were also tested under physiological oxygen levels. RESULTS: STF-62247 specifically induces autophagic cell death in cells that have lost VHL, an essential mutation in the development of RCC. Treatment with STF-62247 did not alter cell cycle progression but when combined with radiation increased cell killing under oxic and hypoxic/physiological conditions. CONCLUSIONS: This study highlights the possibility of combining targeted therapeutics such as STF-62247 or temsirolimus with radiation to reduce the reliance on partial or full nephrectomy and improve patient prognosis.


Asunto(s)
Autofagia , Carcinoma de Células Renales/radioterapia , Neoplasias Renales/radioterapia , Tolerancia a Radiación , Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/fisiopatología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/efectos de la radiación , Hipoxia de la Célula , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Humanos , Neoplasias Renales/genética , Neoplasias Renales/fisiopatología , Mutación , Piridinas/farmacología , Dosis de Radiación , Tolerancia a Radiación/fisiología , Sirolimus/análogos & derivados , Sirolimus/farmacología , Tiazoles/farmacología , Ensayo de Tumor de Célula Madre , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA