Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
Sci Transl Med ; 3(94): 94ra70, 2011 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-21813754

RESUMEN

Identifying new targeted therapies that kill tumor cells while sparing normal tissue is a major challenge of cancer research. Using a high-throughput chemical synthetic lethal screen, we sought to identify compounds that exploit the loss of the von Hippel-Lindau (VHL) tumor suppressor gene, which occurs in about 80% of renal cell carcinomas (RCCs). RCCs, like many other cancers, are dependent on aerobic glycolysis for ATP production, a phenomenon known as the Warburg effect. The dependence of RCCs on glycolysis is in part a result of induction of glucose transporter 1 (GLUT1). Here, we report the identification of a class of compounds, the 3-series, exemplified by STF-31, which selectively kills RCCs by specifically targeting glucose uptake through GLUT1 and exploiting the unique dependence of these cells on GLUT1 for survival. Treatment with these agents inhibits the growth of RCCs by binding GLUT1 directly and impeding glucose uptake in vivo without toxicity to normal tissue. Activity of STF-31 in these experimental renal tumors can be monitored by [(18)F]fluorodeoxyglucose uptake by micro-positron emission tomography imaging, and therefore, these agents may be readily tested clinically in human tumors. Our results show that the Warburg effect confers distinct characteristics on tumor cells that can be selectively targeted for therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Células Renales/tratamiento farmacológico , Transportador de Glucosa de Tipo 1/metabolismo , Neoplasias Renales/tratamiento farmacológico , Adenosina Trifosfato/biosíntesis , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis , Carcinoma de Células Renales/metabolismo , Glucosa/metabolismo , Glucólisis , Humanos , Neoplasias Renales/metabolismo
9.
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
10.
Nat Rev Drug Discov ; 10(5): 351-64, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21532565

RESUMEN

Unique features of tumours that can be exploited by targeted therapies are a key focus of current cancer research. One such approach is known as synthetic lethality screening, which involves searching for genetic interactions of two mutations whereby the presence of either mutation alone has no effect on cell viability but the combination of the two mutations results in cell death. The presence of one of these mutations in cancer cells but not in normal cells can therefore create opportunities to selectively kill cancer cells by mimicking the effect of the second genetic mutation with targeted therapy. Here, we summarize strategies that can be used to identify synthetic lethal interactions for anticancer drug discovery, describe examples of such interactions that are currently being investigated in preclinical and clinical studies of targeted anticancer therapies, and discuss the challenges of realizing the full potential of such therapies.


Asunto(s)
Antineoplásicos/farmacología , Sistemas de Liberación de Medicamentos , Neoplasias/tratamiento farmacológico , Animales , Supervivencia Celular/genética , Diseño de Fármacos , Descubrimiento de Drogas , Ensayos de Selección de Medicamentos Antitumorales , Genes Letales , Genes Sintéticos , Humanos , Neoplasias/genética
11.
Mol Biol Cell ; 21(18): 3247-57, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20660157

RESUMEN

Hypoxia-inducible factor-1 (HIF-1) is a key transcription factor for responses to low oxygen. Different nonhypoxic stimuli, including hormones and growth factors, are also important HIF-1 activators in the vasculature. Angiotensin II (Ang II), the main effecter hormone in the renin-angiotensin system, is a potent HIF-1 activator in vascular smooth muscle cells (VSMCs). HIF-1 activation by Ang II involves intricate mechanisms of HIF-1α transcription, translation, and protein stabilization. Additionally, the generation of reactive oxygen species (ROS) is essential for HIF-1 activation during Ang II treatment. However, the role of the different VSMC ROS generators in HIF-1 activation by Ang II remains unclear. This work aims at elucidating this question. Surprisingly, repression of NADPH oxidase-generated ROS, using Vas2870, a specific inhibitor or a p22(phox) siRNA had no significant effect on HIF-1 accumulation by Ang II. In contrast, repression of mitochondrial-generated ROS, by complex III inhibition, by Rieske Fe-S protein siRNA, or by the mitochondrial-targeted antioxidant SkQ1, strikingly blocked HIF-1 accumulation. Furthermore, inhibition of mitochondrial-generated ROS abolished HIF-1α protein stability, HIF-1-dependent transcription and VSMC migration by Ang II. A large number of studies implicate NADPH oxidase-generated ROS in Ang II-mediated signaling pathways in VSMCs. However, our work points to mitochondrial-generated ROS as essential intermediates for HIF-1 activation in nonhypoxic conditions.


Asunto(s)
Factor 1 Inducible por Hipoxia/metabolismo , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Angiotensina II/farmacología , Animales , Antibacterianos/farmacología , Antifúngicos/farmacología , Células Cultivadas , Complejo III de Transporte de Electrones/genética , Complejo III de Transporte de Electrones/metabolismo , Masculino , Metacrilatos/farmacología , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , NADPH Oxidasas/metabolismo , Polienos/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Wistar , Tiazoles/farmacología
12.
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
13.
J Med Chem ; 53(2): 787-97, 2010 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-19994864

RESUMEN

Renal cell carcinomas (RCC) are refractory to standard therapy with advanced RCC having a poor prognosis; consequently treatment of advanced RCC represents an unmet clinical need. The von Hippel-Lindau (VHL) tumor suppressor gene is mutated or inactivated in a majority of RCCs. We recently identified a 4-pyridyl-2-anilinothiazole (PAT) with selective cytotoxicity against VHL-deficient renal cells mediated by induction of autophagy and increased acidification of autolysosomes. We report exploration of structure-activity relationships (SAR) around this PAT lead. Analogues with substituents on each of the three rings, and various linkers between rings, were synthesized and tested in vitro using paired RCC4 cell lines. A contour map describing the relative spatial contributions of different chemical features to potency illustrates a region, adjacent to the pyridyl ring, with potential for further development. Examples probing this domain validated this approach and may provide the opportunity to develop this novel chemotype as a targeted approach to the treatment of RCC.


Asunto(s)
Compuestos de Anilina/farmacología , Autofagia/efectos de los fármacos , Carcinoma de Células Renales/tratamiento farmacológico , Tiazoles/farmacología , Enfermedad de von Hippel-Lindau/tratamiento farmacológico , Compuestos de Anilina/química , Compuestos de Anilina/uso terapéutico , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Humanos , Lisosomas/efectos de los fármacos , Piridinas , Relación Estructura-Actividad , Tiazoles/química , Tiazoles/uso terapéutico , Enfermedad de von Hippel-Lindau/patología
14.
Cancer Cell ; 15(6): 527-38, 2009 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-19477431

RESUMEN

Sustained angiogenesis, through either local sprouting (angiogenesis) or the recruitment of bone marrow-derived cells (BMDCs) (vasculogenesis), is essential to the development of a tumor. How BMDCs are recruited to the tumor and their contribution to the tumor vasculature is poorly understood. Here, we demonstrate that both IL-8 and angiogenin contribute to the complementary pathways of angiogenesis and BMDC mobilization to increase tumor growth. These two factors are regulated by PHD2 in a HIF-independent but NF-kappaB-dependent manner. PHD2 levels are decreased in human cancers, compared with corresponding normal tissue, and correlate with an increase in mature blood vessels. Thus, PHD2 plays a critical role in regulating tumor angiogenesis.


Asunto(s)
Células de la Médula Ósea/fisiología , Endotelio Vascular/patología , Neoplasias Experimentales/irrigación sanguínea , Neovascularización Patológica/metabolismo , Procolágeno-Prolina Dioxigenasa/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células Cultivadas , Células Endoteliales/patología , Células Endoteliales/fisiología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Interleucina-8/metabolismo , Masculino , Ratones , Ratones SCID , FN-kappa B/metabolismo , Trasplante de Neoplasias , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Neovascularización Patológica/patología , Ribonucleasa Pancreática/metabolismo
15.
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
16.
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
17.
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
18.
Cell Cycle ; 7(19): 2987-90, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18818511

RESUMEN

Standard cytotoxic agents for treating cancer were developed based on their effectiveness to kill rapidly dividing cells, not on their ability to selectively kill cancer cells and spare normal tissue. Much of contemporary cancer research is aimed at identifying specific molecular features of cancers to directly target tumor cells with the hope of reducing or eliminating unwanted side effects. Targeted therapy for the treatment of cancer can be divided into two main categories: monoclonal antibodies and small molecules. In this Perspective, we review the approach of synthetic lethality to target cancer, specifically renal cell carcinoma. The concept of synthetic lethality is used to describe a genetic interaction of two non-allelic and non-lethal genes that when mutated simultaneously results in cell death. Recently, we identified a compound, STF-62247, that functions in a synthetic lethal manner to the loss of VHL, a mutation found in the majority of renal cell carcinomas.


Asunto(s)
Autofagia/genética , Genes Letales/genética , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Animales , Humanos , Modelos Biológicos , Proteínas Proto-Oncogénicas c-ret/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-ret/metabolismo , Piridinas/farmacología , Tiazoles/farmacología , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
19.
Cancer Cell ; 14(1): 90-102, 2008 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-18598947

RESUMEN

Renal cell carcinomas (RCCs) are refractory to standard therapies. The von Hippel-Lindau (VHL) tumor suppressor gene is inactivated in 75% of RCCs. By screening for small molecules selectively targeting VHL-deficient RCC cells, we identified STF-62247. STF-62247 induces cytotoxicity and reduces tumor growth of VHL-deficient RCC cells compared to genetically matched cells with wild-type VHL. STF-62247-stimulated toxicity occurs in a HIF-independent manner through autophagy. Reduction of protein levels of essential autophagy pathway components reduces sensitivity of VHL-deficient cells to STF-62247. Using a yeast deletion pool, we show that loss of proteins involved in Golgi trafficking increases killing by STF-62247. Thus, we have found a small molecule that selectively induces cell death in VHL-deficient cells, representing a paradigm shift for targeted therapy.


Asunto(s)
Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Carcinoma de Células Renales/patología , Neoplasias Renales/patología , Piridinas/farmacología , Tiazoles/farmacología , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Animales , Antineoplásicos/química , Proteína 5 Relacionada con la Autofagia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinoma de Células Renales/enzimología , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Aparato de Golgi/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Factor 1 Inducible por Hipoxia/metabolismo , Concentración 50 Inhibidora , Neoplasias Renales/enzimología , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Masculino , Ratones , Ratones SCID , Proteínas Asociadas a Microtúbulos/metabolismo , Estructura Molecular , Fosfatidilinositol 3-Quinasas/metabolismo , Transporte de Proteínas , Relación Estructura-Actividad , Factores de Tiempo , Transfección , Vacuolas/efectos de los fármacos , Vacuolas/metabolismo , Vacuolas/patología , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Levaduras/efectos de los fármacos , Levaduras/crecimiento & desarrollo , Levaduras/metabolismo
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA