Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Angiogenesis ; 19(3): 257-73, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27146677

RESUMEN

Aberrant activation of the hypoxia inducible factor (HIF) pathway causing overexpression of angiogenic genes, like vascular endothelial growth factor (VEGF), is one of the underlying causes of ocular neovascularization (NV) and metastatic cancer. Consistently, along with surgical interventions, a number of anti-VEGF agents have been approved by FDA for the treatment of ocular neovascular diseases. These anti-VEGF agents, like ranibizumab/lucentis, have revolutionized the treatment in the past decade. However, substantial vision improvement is observed only in a subset of age-related macular degeneration patients receiving ranibizumab. Further, all current therapies are associated with limitations and side effects. For example, surgeries cause tissue destruction and inflammation while anti-VEGF therapies are expensive, require repeated administration, and offer temporary relief from vascular leakage. These factors impose significant cost and treatment burdens to both the patient and society. With an aging population in most western countries with a continually increasing number of patients on lifelong treatment for these retinal diseases, the focus of ocular drug development for neovascular diseases will be to improve efficacy while reducing treatment costs. Blocking the HIF pathway, a major regulator of ocular NV and cancer, offers an appealing therapeutic strategy. Therefore, this review summarizes HIF inhibitors that have been recently evaluated for the treatment of different cancers and ischemic retinopathies.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Retiniana/tratamiento farmacológico , Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Inductores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Antraciclinas/uso terapéutico , Compuestos de Bifenilo/uso terapéutico , Glicósidos Cardíacos/uso terapéutico , Humanos , Indazoles/uso terapéutico , Lignanos/uso terapéutico , Modelos Biológicos
2.
Biochem Biophys Res Commun ; 438(1): 71-7, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23872148

RESUMEN

Hemangioblastomas of the retina, central nervous system, and kidney are observed in patients with mutations in the von Hippel-Lindau (VHL) tumor suppressor gene. Mutations in the VHL lead to constitutive activation of hypoxia-inducible-factor (HIF) pathway. HIF-mediated expression of pro-angiogenic genes causes extensive pathological neovascularization in hemangioblastomas. A number of studies have shown coexistence of pro-angiogenic and stem cell markers in 'tumorlet-like stromal cells' in the retinal and optic nerve hemangioblastomas, leading to suggestions that hemangioblastomas originate from developmentally arrested stem cells or embryonic progenitors. Since recent studies have shown that the HIF pathway also plays a role in the maintenance/de-differentiation of normal and cancerous stem cells, we evaluated the role of the HIF pathway in the expression of stem cell markers in VHL-/- renal cell carcinoma cells under normoxia or VHL+/+ retinal pigment epithelial cells under hypoxia. Here we show that the expression of stem cell markers in hemangioblastomas is due to activation of the HIF pathway. Further, we show that honokiol, digoxin, and doxorubicin, three recently identified HIF inhibitors from natural sources, blocks the expression of stem cell markers. Our results show the mechanism for the cytological origin of neoplastic stromal cells in hemangioblastomas, and suggest that inhibition of the HIF pathway is an attractive strategy for the treatment of hemangioblastomas.


Asunto(s)
Hemangioblastoma/metabolismo , Hemangioblastoma/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Hipoxia de la Célula , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos
3.
Biochem Biophys Res Commun ; 438(4): 697-702, 2013 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-23921228

RESUMEN

Aberrant activation of the hypoxia inducible factor (HIF) pathway is the underlying cause of retinal neovascularization, one of the most common causes of blindness worldwide. The HIF pathway also plays critical roles during tumor angiogenesis and cancer stem cell transformation. We have recently shown that honokiol is a potent inhibitor of the HIF pathway in a number of cancer and retinal pigment epithelial cell lines. Here we evaluate the safety and efficacy of honokiol, digoxin, and doxorubicin, three recently identified HIF inhibitors from natural sources. Our studies show that honokiol has a better safety to efficacy profile as a HIF inhibitor than digoxin and doxorubicin. Further, we show for the first time that daily intraperitoneal injection of honokiol starting at postnatal day (P) 12 in an oxygen-induced retinopathy (OIR) mouse model significantly reduced retinal neovascularization at P17. Administration of honokiol also prevents the oxygen-induced central retinal vaso-obliteration, characteristic feature of the OIR model. Additionally, honokiol enhanced physiological revascularization of the retinal vascular plexuses. Since honokiol suppresses multiple pathways activated by HIF, in addition to the VEGF signaling, it may provide advantages over current treatments utilizing specific VEGF antagonists for ocular neovascular diseases and cancers.


Asunto(s)
Compuestos de Bifenilo/uso terapéutico , Medicamentos Herbarios Chinos/uso terapéutico , Lignanos/uso terapéutico , Retina/efectos de los fármacos , Retina/patología , Neovascularización Retiniana/tratamiento farmacológico , Neovascularización Retiniana/patología , Animales , Antibióticos Antineoplásicos/uso terapéutico , Línea Celular , Digoxina/uso terapéutico , Doxorrubicina/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Humanos , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Endogámicos C57BL , Oxígeno , Retina/metabolismo , Neovascularización Retiniana/inducido químicamente , Neovascularización Retiniana/genética , Activación Transcripcional/efectos de los fármacos
4.
Biochem Biophys Res Commun ; 422(3): 369-74, 2012 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-22580280

RESUMEN

Hypoxia-inducible-factor (HIF)-mediated expression of pro-angiogenic genes under hypoxic conditions is the fundamental cause of pathological neovascularization in retinal ischemic diseases and cancers. Recent studies have shown that histone lysine demethylases (KDMs) play a key role in the amplification of HIF signaling and expression of pro-angiogenic genes. Thus, the inhibitors of the HIF pathway or KDMs can have profound therapeutic value for diseases caused by pathological neovascularization. Here, we show that hypoxia-mediated expression of KDMs is a conserved process across multiple cell lines. Moreover, we report that honokiol, a biphenolic phytochemical extracted from Magnolia genus which has been used for thousands of years in the traditional Japanese and Chinese medicine, is a potent inhibitor of the HIF pathway as well as hypoxia-induced expression of KDMs in a number of cancer and retinal pigment epithelial cell lines. Further, treating the cells with honokiol leads to inhibition of KDM-mediated induction of pro-angiogenic genes (adrenomedullin and growth differentiation factor 15) under hypoxic conditions. Our results provide an evidence-based scientific explanation for therapeutic benefits observed with honokiol and warrant its further clinical evaluation for the treatment of pathological neovascularization in retinal ischemic diseases and cancers.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Compuestos de Bifenilo/farmacología , Histona Demetilasas/antagonistas & inhibidores , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Lignanos/farmacología , Neovascularización Patológica/metabolismo , Hipoxia de la Célula , Línea Celular , Línea Celular Tumoral , Expresión Génica/efectos de los fármacos , Humanos , Hipoxia/metabolismo , Neovascularización Patológica/genética , Oxígeno/metabolismo
5.
Biochem Biophys Res Commun ; 405(4): 588-92, 2011 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-21266164

RESUMEN

Jumonji domain containing iron (II), 2-oxoglutarate (2OG)-dependent dioxygenases from Jmjd2 family demethylate trimethylated histone3-lysine 9 (H3-K9me3), and also H3-K9me2 and H3-K36me3, albeit at lower rates. Recently, we have identified the first non-histone substrates of JmjD2 demethylases. Here, we studied the substrate specificity of Jmjd2a-c demethylases using site-directed mutagenesis and novel non-histone substrates. We identified preference of Arg at -1 position and a smaller amino acid at -2 position using both singly and doubly mutated peptide substrates by Jmjd2a-c demethylases. Our results also identified similarities in substrate selectivity by H3-K9 methyltransferase, G9a and Jmjd2 demethylases despite their distinct reaction mechanisms.


Asunto(s)
Histona Demetilasas con Dominio de Jumonji/química , Arginina/química , Arginina/genética , Clonación Molecular , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Mutagénesis Sitio-Dirigida , Especificidad por Sustrato
6.
Biochem Biophys Res Commun ; 415(2): 373-7, 2011 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-22037463

RESUMEN

Hypoxia inducible factor (HIF) plays a critical role in cellular adaptation to hypoxia by regulating the expression of essential genes. Pathological activation of this pathway leads to the expression of pro-angiogenic factors during the neovascularization in cancer and retinal diseases. Little is known about the epigenetic regulations during HIF-mediated transcription and activation of pro-angiogenic genes in oxygen-dependent retinal diseases. Here, we show that hypoxia induces the expression of a number of histone lysine demethylases (KDMs) in retinal pigment epithelial cells. Moreover, we show that the expression of pro-angiogenic genes (ADM, GDF15, HMOX1, SERPE1 and SERPB8) is dependent on KDMs under hypoxic conditions. Further, treating the cells with a general KDM inhibitor blocks the expression of these pro-angiogenic genes. Results from these studies identify a new layer of epigenetic transcription regulation under hypoxic conditions and suggest that specific inhibitors of KDMs such as JMJD1A can be a new therapeutic approach to treat diseases caused by the hypoxia induced neovascularization in cancer and retinal diseases.


Asunto(s)
Epigénesis Genética , Histona Demetilasas/biosíntesis , Oxígeno/metabolismo , Neovascularización Retiniana/enzimología , Neovascularización Retiniana/genética , Epitelio Pigmentado de la Retina/enzimología , Adrenomedulina/genética , Aminoácidos Dicarboxílicos/farmacología , Hipoxia de la Célula/genética , Línea Celular Tumoral , Factor 15 de Diferenciación de Crecimiento/genética , Hemo-Oxigenasa 1/genética , Histona Demetilasas/genética , Humanos , Epitelio Pigmentado de la Retina/efectos de los fármacos
7.
Biochem Biophys Res Commun ; 390(2): 280-4, 2009 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-19799855

RESUMEN

Recent studies have shown that some Jumonji domain containing proteins demethylate tri- and dimethylated histone lysines by catalyzing a dioxygenase reaction. Here we report the substrate specificity of Jumonji domain-2 family histone demethylases (JMJD2A-C). A candidate substrate-based approach demonstrated that in addition to its known substrate, trimethylated histone H3-lysine-9, JMJD2A-C demethylate trimethylated lysine containing peptides from WIZ, CDYL1, CSB and G9a proteins, all constituents of transcription repression complexes. Our results are consistent with lax substrate specificities observed for the iron (II), 2-oxoglutarate-dependent dioxygenases, and shed new light on signaling pathways regulated by Jumonji domain-2 family histone demethylases during epigenetic transcriptional regulation.


Asunto(s)
Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Secuencia de Aminoácidos , Histonas/química , Humanos , Histona Demetilasas con Dominio de Jumonji/química , Histona Demetilasas con Dominio de Jumonji/genética , Lisina/química , Lisina/metabolismo , Datos de Secuencia Molecular , Transducción de Señal , Especificidad por Sustrato
8.
PLoS One ; 9(11): e113717, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25422886

RESUMEN

Pathological activation of the hypoxia-inducible-factor (HIF) pathway leading to expression of pro-angiogenic genes, such as vascular endothelial growth factor (VEGF), is the fundamental cause of neovascularization in ocular ischemic diseases and cancers. We have shown that pure honokiol inhibits the HIF pathway and hypoxia-mediated expression of pro-angiogenic genes in a number of cancer and retinal pigment epithelial (RPE) cell lines. The crude extracts, containing honokiol, from Magnolia plants have been used for thousands of years in the traditional oriental medicine for a number of health benefits. We have recently demonstrated that daily intraperitoneal injection of honokiol starting at postnatal day (P) 12 in an oxygen induced retinopathy mouse model significantly reduced retinal neovascularization at P17. Here, we evaluate the mechanism of HIF inhibition by honokiol in RPE cells. Using chromatin immunoprecipitation experiments, we demonstrate that honokiol inhibits binding of HIF to hypoxia-response elements present on VEGF promoter. We further show using a number of in vitro angiogenesis assays that, in addition to anti-HIF effect, honokiol manifests potent anti-angiogenic effect on human retinal micro vascular endothelial cells. Our results suggest that honokiol possesses potent anti-HIF and anti-angiogenic properties. These properties of honokiol make it an ideal therapeutic agent for the treatment of ocular neovascular diseases and solid tumors.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Compuestos de Bifenilo/uso terapéutico , Oftalmopatías/tratamiento farmacológico , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Lignanos/uso terapéutico , Neovascularización Patológica/tratamiento farmacológico , Inhibidores de la Angiogénesis/farmacología , Compuestos de Bifenilo/farmacología , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Células HeLa , Humanos , Lignanos/farmacología , Luciferasas/antagonistas & inhibidores , Epitelio Pigmentado de la Retina/citología
9.
Toxicol Rep ; 1: 1152-1161, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-28962325

RESUMEN

BACKGROUND: Epigenetic modifications, particularly DNA methylation and posttranslational histone modifications regulate heritable changes in transcription without changes in the DNA sequence. Despite a number of studies showing clear links between environmental factors and DNA methylation, little is known about the effect of environmental factors on the recently identified histone lysine methylation. Since their identification numerous studies have establish critical role played by these enzymes in mammalian development. OBJECTIVES: Identification of the Jumonji (Jmj) domain containing histone lysine demethylase have added a new dimension to epigenetic control of gene expression by dynamic regulation of histone methylation marks. The objective of our study was to evaluate the effect of prohexadione and trinexapac, widely used plant growth regulators of the acylcyclohexanediones class, on the enzymatic activity of histone lysine demethylases and histone modifications during the neural stem/progenitor cell differentiation. METHODS: Here we show that prohexadione, but not trinexapac, directly inhibits non-heme iron (II), 2-oxoglutarate-dependent histone lysine demethylase such as Jmjd2a. We used molecular modeling to show binding of prohexadione to Jmjd2a. We also performed in vitro demethylation assays to show the inhibitory effect of prohexadione on Jmjd2a. Further we tested this molecule in cell culture model of mouse hippocampal neural stem/progenitor cells to demonstrate its effect toward neuronal proliferation and differentiation. RESULTS: Molecular modeling studies suggest that prohexadione binds to the 2-oxoglutarate binding site of Jmjd2a demethylase. Treatment of primary neural stem/progenitor cells with prohexadione showed a concentration dependent reduction in their proliferation. Further, the prohexadione treated neurospheres were induced toward neurogenic lineage upon differentiation. CONCLUSIONS: Our results describe an important chemico-biological interaction of prohexadione, in light of critical roles played by histone lysine demethylases in human health and diseases.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA