Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 240
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Mar Drugs ; 22(4)2024 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-38667760

RESUMEN

The inadequate vascularization seen in fast-growing solid tumors gives rise to hypoxic areas, fostering specific changes in gene expression that bolster tumor cell survival and metastasis, ultimately leading to unfavorable clinical prognoses across different cancer types. Hypoxia-inducible factors (HIF-1 and HIF-2) emerge as druggable pivotal players orchestrating tumor metastasis and angiogenesis, thus positioning them as prime targets for cancer treatment. A range of HIF inhibitors, notably natural compounds originating from marine organisms, exhibit encouraging anticancer properties, underscoring their significance as promising therapeutic options. Bioprospection of the marine environment is now a well-settled approach to the discovery and development of anticancer agents that might have their medicinal chemistry developed into clinical candidates. However, despite the massive increase in the number of marine natural products classified as 'anticancer leads,' most of which correspond to general cytotoxic agents, and only a few have been characterized regarding their molecular targets and mechanisms of action. The current review presents a critical analysis of inhibitors of HIF-1 and HIF-2 and hypoxia-selective compounds that have been sourced from marine organisms and that might act as new chemotherapeutic candidates or serve as templates for the development of structurally similar derivatives with improved anticancer efficacy.


Asunto(s)
Antineoplásicos , Organismos Acuáticos , Productos Biológicos , Factor 1 Inducible por Hipoxia , Neoplasias , Animales , Humanos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Organismos Acuáticos/química , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Productos Biológicos/farmacología , Productos Biológicos/química , Productos Biológicos/uso terapéutico , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Transducción de Señal/efectos de los fármacos
2.
J Biochem Mol Toxicol ; 36(2): e22954, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34783123

RESUMEN

Marine resources are notably explored for their unique biomolecules that have been designed to be drug targets for their immense potential against various pathologies. These biomolecules are mostly secondary metabolites from different species that include sponges, tunicates, echinoderms, ascidians, algae, and marine symbionts. Among the various biological activities of the marine biomolecules, antiangiogenic property has gained much significance in alternate therapy for treatment against cancer. Hypoxia inducible factor (HIF) and vascular endothelial growth factor (VEGF) are the prime signaling pathways related to angiogenesis that are exclusively designated as markers for critical selection of novel inhibitors. This is mainly due to their importance in tumor induction and regulatory control over other interlinked pathways involved in cancer. Small molecular drug screening using the zebrafish model has been an advantage in cancer research in recent times. This review addresses the importance of marine biomolecules and their antiangiogenic efficacy by targeting HIF/VEGF pathways experimented in the zebrafish model in the last decade. Thus, it would provide more clear insights into the role of biomolecules in alternative cancer therapy.


Asunto(s)
Inhibidores de la Angiogénesis , Organismos Acuáticos/química , Endotelio Vascular/metabolismo , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Neovascularización Patológica/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Proteínas de Pez Cebra/antagonistas & inhibidores , Pez Cebra/metabolismo , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/farmacología , Animales , Modelos Animales de Enfermedad , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , Neovascularización Patológica/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas de Pez Cebra/metabolismo
3.
Pharmacol Res ; 170: 105742, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34182129

RESUMEN

Stroke is an acute cerebrovascular disease caused by sudden rupture of blood vessels in the brain or blockage of blood vessels, which has now become one of the main causes of adult death. During stroke, hypoxia-inducible factor-1 (HIF-1), as an important regulator under hypoxia conditions, is involved in the pathological process of stroke by regulating multi-pathways, such as glucose metabolism, angiogenesis, erythropoiesis, cell survival. However, the roles of HIF-1 in stroke are still controversial, which are related with ischemic time and degree of ischemia. The regulatory mechanisms of HIF-1 in stroke include inflammation, autophagy, oxidative stress, apoptosis and energy metabolism. The potential drugs targeting HIF-1 have attracted more attention, such as HIF-1 inhibitors, HIF-1 stabilizers and natural products. Based on the role of HIF-1 in stroke, HIF-1 is expected to be a potential target for stroke treatment. Resolving when and what interventions for HIF-1 to take during stroke will provide novel strategies for stroke treatment.


Asunto(s)
Encéfalo/efectos de los fármacos , Diseño de Fármacos , Desarrollo de Medicamentos , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Terapia Molecular Dirigida , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Encéfalo/metabolismo , Encéfalo/fisiopatología , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , Estabilidad Proteica , Transducción de Señal , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/fisiopatología
4.
Cancer Control ; 28: 10732748211041243, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34554006

RESUMEN

In the 1920s, Otto Warburg observed the phenomenon of altered glucose metabolism in cancer cells. Although the initial hypothesis suggested that the alteration resulted from mitochondrial damage, multiple studies of the subject revealed a precise, multistage process rather than a random pattern. The phenomenon of aerobic glycolysis emerges not only from mitochondrial abnormalities common in cancer cells, but also results from metabolic reprogramming beneficial for their sustenance. The Warburg effect enables metabolic adaptation of cancer cells to grow and proliferate, simultaneously enabling their survival in hypoxic conditions. Altered glucose metabolism of cancer cells includes, inter alia, qualitative and quantitative changes within glucose transporters, enzymes of the glycolytic pathway, such as hexokinases and pyruvate kinase, hypoxia-inducible factor, monocarboxylate transporters, and lactate dehydrogenase. This review summarizes the current state of knowledge regarding inhibitors of cancer glucose metabolism with a focus on their clinical potential. The altered metabolic phenotype of cancer cells allows for targeting of specific mechanisms, which might improve conventional methods in anti-cancer therapy. However, several problems such as drug bioavailability, specificity, toxicity, the plasticity of cancer cells, and heterogeneity of cells in tumors have to be overcome when designing therapies based on compounds targeted in cancer cell energy metabolism.


Asunto(s)
Glucólisis/fisiología , Neoplasias/fisiopatología , Efecto Warburg en Oncología , Antineoplásicos/farmacología , Humanos , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , L-Lactato Deshidrogenasa/biosíntesis , Transportadores de Ácidos Monocarboxílicos/biosíntesis
5.
Bioorg Chem ; 109: 104740, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33626453

RESUMEN

Six new non-classical cardenolides (1-6), and seventeen known ones (7-23) were isolated from Calotropis gigantea. All cardenolides showed inhibitory effect on hypoxia inducible factor-1 (HIF-1) transcriptional activity with IC50 of 8.85 nM-16.69 µM except 5 and 7. The novel 19-dihydrocalotoxin (1) exhibited a comparable HIF-1 inhibitory activity (IC50 of 139.57 nM) to digoxin (IC50 of 145.77 nM), a well-studied HIF-1 inhibitor, and 11, 12, 14, 16 and 19 presented 1.4-15.4 folds stronger HIF-1 inhibition than digoxin. 1 and 11 showed a dose-dependent inhibition on HIF-1α protein, which led to their HIF-1 suppressing effects. Compared with LO2 and H9c2 normal cell lines, both 1 and 11 showed selective cytotoxicity against various cancer cell lines including HCT116, HeLa, HepG2, A549, MCF-7, A2780 and MDA-MB-231. Moreover, a comprehensive structure-activity relationship was concluded for these non-classical cardenolides as HIF-1 inhibitors, which may shed some light on the rational design and development of cardenolide-based anticancer drugs.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Calotropis/química , Cardenólidos/farmacología , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Extractos Vegetales/farmacología , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/aislamiento & purificación , Cardenólidos/química , Cardenólidos/aislamiento & purificación , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Conformación Molecular , Extractos Vegetales/química , Extractos Vegetales/aislamiento & purificación , Relación Estructura-Actividad
6.
J Pharmacol Sci ; 144(4): 229-236, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33070842

RESUMEN

The kidneys are the major organs for erythropoietin (EPO) production in adults, and thus, kidney damage results in reduced EPO levels and anemia. Inhibitors of Hypoxia-inducible factor-prolyl hydroxylase domain-containing protein (HIF-PHD) are awaited as new therapeutic options for renal anemia. It can be predicted that most patients who receive HIF-PHD inhibitors have renal dysfunction as a cause of anemia. Therefore, in the present study, we investigated the effects of the HIF-PHD inhibitor molidustat on anemia and renal dysfunction when initiated after the onset of renal anemia. Male C57BL/6J mice received adenine orally to induce nephropathy. After the onset of nephropathy, the mice were treated with either vehicle or molidustat. After 4 weeks of administration, vehicle-treated mice displayed significant anemia, and molidustat ameliorated this anemia. Vehicle-treated mice exhibited reduced creatinine clearance and body weight, increased blood urea nitrogen levels, histopathological changes, immune cell infiltration, and dehydration. Molidustat reversed immune cell infiltration, dehydration, and renal fibrosis without improving renal functional parameters. In conclusion, molidustat treatment initiated after the onset of nephropathy and renal anemia reversed anemia in mice. Molidustat improved some parameters of renal abnormality, but it did not restore renal function.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/complicaciones , Adenina/efectos adversos , Anemia/tratamiento farmacológico , Anemia/etiología , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Inhibidores de Prolil-Hidroxilasa , Pirazoles/uso terapéutico , Triazoles/uso terapéutico , Lesión Renal Aguda/metabolismo , Animales , Modelos Animales de Enfermedad , Eritropoyetina/metabolismo , Masculino , Ratones Endogámicos C57BL , Pirazoles/administración & dosificación , Pirazoles/farmacología , Triazoles/administración & dosificación , Triazoles/farmacología
7.
Am J Physiol Renal Physiol ; 317(5): F1265-F1273, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31588798

RESUMEN

Muscle wasting and diminished physical performance contribute to the morbidity and mortality of chronic kidney disease (CKD), for which no curative therapy exists. Accumulating evidence indicates that impaired angiogenesis occurs in the muscles of CKD models. Therefore, proangiogenesis therapy is considered a potentially effective strategy for limiting CKD-associated myopathy. Hypoxia-inducible factor (HIF)-prolyl hydroxylase inhibitor (HIF-PHI) stabilizes HIF and enhances muscle angiogenesis during acute ischemia; however, little evidence was available from CKD models. Here, we assessed whether pharmacological activation of HIF by MK-8617 (MK), a novel orally active HIF-PHI, improves CKD-associated myopathy. Mice were divided into sham or CKD groups, and CKD mice were subdivided into CKD + vehicle or MK treatment groups (1.5, 5, or 12.5 mg/kg for 12 wk). In CKD mice, skeletal muscle mass, mitochondrial amount, and exercise capacity decreased compared with sham mice. Compared with the CKD + vehicle group, low (1.5 mg/kg) and medium (5 mg/kg) doses of MK, but not the high dose (12.5 mg/kg), significantly restored these changes and was accompanied by incremental increases in HIF-1α. Furthermore, increased capillary density and area were observed in a MK dose-dependent manner, which is likely related to an improved VEGF response in the skeletal muscle of CKD mice. In addition, macrophage and proinflammatory cytokines, including monocyte chemoattractant protein 1, TNF-α, and IL-6, significantly increased in the high-dose MK group. These results indicate that HIF-PHI provides a potential therapeutic strategy to improve CKD-associated myopathy.


Asunto(s)
Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Enfermedades Musculares/tratamiento farmacológico , Enfermedades Musculares/etiología , Inhibidores de Prolil-Hidroxilasa/farmacología , Piridazinas/farmacología , Pirimidinas/farmacología , Insuficiencia Renal Crónica/complicaciones , Administración Oral , Animales , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Fuerza Muscular/efectos de los fármacos , Músculo Esquelético/patología , Enfermedades Musculares/patología , Piridazinas/administración & dosificación , Pirimidinas/administración & dosificación
8.
Kidney Int ; 95(1): 50-56, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30606429

RESUMEN

Renal ischemia-reperfusion injury (IRI) is a significant clinical challenge faced by clinicians perioperatively in kidney transplantation. Recent work has demonstrated the key importance of transmembrane receptors in the injured tubular epithelial cell, most notably Toll-like receptors, activated by exogenous and endogenous ligands in response to external and internal stresses. Through sequential protein-protein interactions, the signal is relayed deep into the core physiological machinery of the cell, having numerous effects from upregulation of pro-inflammatory gene products through to modulating mitochondrial respiration. Inter-pathway cross talk facilitates a co-ordinated response at an individual cellular level, as well as modulating the surrounding tissue's microenvironment through close interactions with the endothelium and circulating leukocytes. Defining the underlying cellular cascades involved in IRI will assist the identification of novel interventional targets to attenuate IRI with the potential to improve transplantation outcomes. We present a focused review of 3 key cellular signalling pathways in the injured tubular epithelial cell that have been the focus of much research over the past 2 decades: toll-like receptors, sphingosine-1-phosphate receptors and hypoxia inducible factors. We provide a unique perspective on the potential clinical translations of this recent work in the transplant setting. This is particularly timely with the recent completion of phase I and ongoing phase 2 clinical trials of inhibitors targeting specific components of these signaling cascades.


Asunto(s)
Células Epiteliales/patología , Trasplante de Riñón/efectos adversos , Túbulos Renales/patología , Daño por Reperfusión/patología , Aloinjertos/patología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Ensayos Clínicos Fase II como Asunto , Humanos , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Factor 1 Inducible por Hipoxia/metabolismo , Fallo Renal Crónico/cirugía , Túbulos Renales/citología , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Receptores de Lisoesfingolípidos/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/etiología , Transducción de Señal/efectos de los fármacos , Receptores Toll-Like/antagonistas & inhibidores , Receptores Toll-Like/metabolismo
9.
Blood ; 130(5): 655-665, 2017 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-28576876

RESUMEN

Chronic myeloid leukemia (CML) is a hematopoietic stem cell (HSC)-driven neoplasia characterized by expression of the constitutively active tyrosine kinase BCR/Abl. CML therapy based on tyrosine kinase inhibitors (TKIs) is highly effective in inducing remission but not in targeting leukemia stem cells (LSCs), which sustain minimal residual disease and are responsible for CML relapse following discontinuation of treatment. The identification of molecules capable of targeting LSCs appears therefore of primary importance to aim at CML eradication. LSCs home in bone marrow areas at low oxygen tension, where HSCs are physiologically hosted. This study addresses the effects of pharmacological inhibition of hypoxia-inducible factor-1 (HIF-1), a critical regulator of LSC survival, on the maintenance of CML stem cell potential. We found that the HIF-1 inhibitor acriflavine (ACF) decreased survival and growth of CML cells. These effects were paralleled by decreased expression of c-Myc and stemness-related genes. Using different in vitro stem cell assays, we showed that ACF, but not TKIs, targets the stem cell potential of CML cells, including primary cells explanted from 12 CML patients. Moreover, in a murine CML model, ACF decreased leukemia development and reduced LSC maintenance. Importantly, ACF exhibited significantly less-severe effects on non-CML hematopoietic cells in vitro and in vivo. Thus, we propose ACF, a US Food and Drug Administration (FDA)-approved drug for nononcological use in humans, as a novel therapeutic approach to prevent CML relapse and, in combination with TKIs, enhance induction of remission.


Asunto(s)
Acriflavina/farmacología , Sistemas de Liberación de Medicamentos/métodos , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Leucemia Mielógena Crónica BCR-ABL Positiva , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias Experimentales , Células Madre Neoplásicas/metabolismo , Animales , Supervivencia Celular , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Ratones , Células 3T3 NIH , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Células Madre Neoplásicas/patología
10.
Bioorg Med Chem Lett ; 29(16): 2327-2331, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31281016

RESUMEN

The novel marine pyrrole alkaloid neolamellarin A derived from sponge has been shown to inhibit hypoxia-induced HIF-1 activity. In this work, we designed and synthesized neolamellarin A and its series of derivatives by a convergent synthetic strategy. The HIF-1 inhibitory activity and cytotoxicity of these compounds were evaluated in Hela cells by dual-luciferase reporter gene assay and MTT assay, respectively. The results showed that neolamellarin A 1 (IC50 = 10.8 ±â€¯1.0 µM) and derivative 2b (IC50 = 11.9 ±â€¯3.6 µM) had the best HIF-1 inhibitory activity and low cytotoxicity. Our SAR research focused on the effects of key regions aliphatic carbon chain length, aromatic ring substituents and C-7 substituent on biological activity, providing a basis for the subsequent research on the development of novel pyrrole alkaloids as HIF-1 inhibitors and design of small molecule probes for target protein identification.


Asunto(s)
Antineoplásicos/farmacología , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Pirroles/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células HeLa , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , Estructura Molecular , Pirroles/síntesis química , Pirroles/química , Relación Estructura-Actividad
11.
Bioorg Chem ; 85: 357-363, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30658235

RESUMEN

In this paper, the mechanism of orobanone analogues formation via aromatization rearrangement of curcumol was minutely explored. Aromatization of curcumol with acetone under acidic condition was selected as the model reaction. The formation of a stable aromatic system was the driving force for this reaction. Based on the model reaction, other four new orobanone analogues were prepared through curcumol reacting with different carbonyl compounds. The results showed that the stability of carbocation, which was generated from the carbonyl compounds, and the steric hindrance were main factors affecting the aromatization. We also synthesized the analogue of aromaticane B using compound 2. In vitro anti-proliferative activity of some derivatives were tested by MTT assay. Two derivatives showed weak anti-tumor effect on two cancer cell lines (HepG2 and MCF7) under normoxia. Four orobanone analogue 2, 5, 6 and 9 significantly inhibited hypoxia-induced HIF-1 luciferase reporter activity in HeLa cells with the IC50 values of 13.6, 6.6, 2.4 and 18.2 µM, respectively.


Asunto(s)
Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Sesquiterpenos/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Factor 1 Inducible por Hipoxia/genética , Sesquiterpenos/síntesis química , Transcripción Genética/efectos de los fármacos
12.
Proc Natl Acad Sci U S A ; 113(18): E2516-25, 2016 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-27091985

RESUMEN

Retinopathy of prematurity (ROP) causes 100,000 new cases of childhood blindness each year. ROP is initiated by oxygen supplementation necessary to prevent neonatal death. We used organ systems pharmacology to define the transcriptomes of mice that were cured of oxygen-induced retinopathy (OIR, ROP model) by hypoxia-inducible factor (HIF) stabilization via HIF prolyl hydroxylase inhibition using the isoquinolone Roxadustat or the 2-oxoglutarate analog dimethyloxalylglycine (DMOG). Although both molecules conferred a protective phenotype, gene expression analysis by RNA sequencing found that Roxadustat can prevent OIR by two pathways: direct retinal HIF stabilization and induction of aerobic glycolysis or indirect hepatic HIF-1 stabilization and increased serum angiokines. As predicted by pathway analysis, Roxadustat rescued the hepatic HIF-1 knockout mouse from retinal oxygen toxicity, whereas DMOG could not. The simplicity of systemic treatment that targets both the liver and the eye provides a rationale for protecting the severely premature infant from oxygen toxicity.


Asunto(s)
Glicina/análogos & derivados , Factor 1 Inducible por Hipoxia/metabolismo , Isoquinolinas/administración & dosificación , Hígado/metabolismo , Retina/metabolismo , Retinopatía de la Prematuridad/tratamiento farmacológico , Retinopatía de la Prematuridad/prevención & control , Transcriptoma/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Glicina/administración & dosificación , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Hígado/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Retina/efectos de los fármacos , Resultado del Tratamiento
13.
J Asian Nat Prod Res ; 20(6): 545-558, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29862843

RESUMEN

Hypoxia-inducible factor-1 (HIF-1), an important transcription factor for tumor survival, is an attractive target for anti-cancer treatment. Herein, we present the design and synthesis of LXY7824, a simplified analogue of 4-O-methylsaucerneol. In addition, its significant HIF-1 inhibitory activity and potent anti-cancer activity in vivo and in vitro were also reported.


Asunto(s)
Antineoplásicos/síntesis química , Diseño de Fármacos , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos
14.
Mol Pharmacol ; 92(5): 510-518, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28814529

RESUMEN

Hypoxia-inducible factor-1 (HIF-1) is a key gene regulator for cellular adaptation to low oxygen. In addition to hypoxia, several nonhypoxic stimuli, including hormones and growth factors, are essential for cell-specific HIF-1 regulation. Our studies have highlighted angiotensin II (AngII), a vasoactive hormone, as a potent HIF-1 activator in vascular smooth muscle cells (VSMC). AngII increases HIF-1 transcriptional activity by modulating specific signaling pathways. In VSMC, p42/p44 mitogen-activated protein kinase (MAPK) pathway activation is essential for HIF-1-mediated transcription during AngII treatment. The present study shows that PD184161, a potent MEK1/2 inhibitor, is an HIF-1 blocker in Ang II-treated VSMC. Unlike PD98059, a widely-used MEK1/2 inhibitor, we found that PD184161 blocked AngII-driven HIF-1α protein induction in a dose-dependent manner. Interestingly, the effect of PD184161 was specific to nonhypoxic activators, since HIF-1α induction by hypoxia (1% O2) was unaffected under similar conditions. VSMC treatment with MG132, a proteasome inhibitor, indicated that PD184161 influenced HIF-1α protein stability. PD184161 also increased HIF-1α binding to von Hippel-Lindau tumor suppressor protein, an E3 ligase component and an indication of HIF-1α hydroxylation. Finally, we show that PD184161 blocked mitochondrial ROS (mtROS) production and cellular ATP levels, at the same time enhancing ascorbate availability in AngII-treated VSMC. Taken together, our study indicates that, independently of p42/p44 MAPK activation, PD184161 blocks mtROS generation by AngII, leading to re-establishment of cellular ascorbate levels, increased VHL binding, and decreased HIF-1α stability. Therefore, this study reveals a previously unsuspected role for PD184161 as an HIF-1 inhibitor in VSMC under nonhypoxic conditions.


Asunto(s)
Compuestos de Anilina/farmacología , Benzamidas/farmacología , Factor 1 Inducible por Hipoxia/fisiología , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Ratas Wistar
15.
Bioorg Med Chem ; 25(6): 1737-1746, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28209257

RESUMEN

Hypoxia-inducible factor-1 (HIF-1) as a key mediator in tumor metastasis, angiogenesis, and poor patient prognosis has been recognized as an important cancer drug target. A novel series of N-(benzofuran-5-yl)aromaticsulfonamide derivatives were synthesized and evaluated as HIF-1 inhibitor. Among these compounds, 7q exhibited specific inhibitory effects on HIF-1 by downregulating the expression of HIF-1α under hypoxic conditions. It inhibited the HIF-1 transcriptional activity (IC50=12.5±0.7µM) and secretion of VEGF (IC50=18.8µM) in MCF-7 cells. Meanwhile, it also significantly suppressed hypoxia-induced migration of HUVEC cells in nontoxic concentrations. Additionally, tube formation assay demonstrated its anti-angiogenesis activity. Finally, the in vivo study indicated that compound 7q could retard angiogenesis in CAM model. These findings supported the HIF-1 inhibitory effect and anti-angiogenic potential of this class of compounds as HIF-1 inhibitor.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Benzofuranos/química , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Sulfonamidas/farmacología , Inhibidores de la Angiogénesis/síntesis química , Inhibidores de la Angiogénesis/química , Células Endoteliales de la Vena Umbilical Humana , Humanos , Sulfonamidas/síntesis química , Sulfonamidas/química , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
Anesth Analg ; 124(6): 1872-1885, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28277320

RESUMEN

Part I of this review discussed the similarities between embryogenesis, mammalian adaptions to hypoxia (primarily driven by hypoxia-inducible factor-1 [HIF-1]), ischemia-reperfusion injury (and its relationship with reactive oxygen species), hibernation, diving animals, cancer, and sepsis, and it focused on the common characteristics that allow cells and organisms to survive in these states. Part II of this review describes techniques by which researchers gain insight into subcellular energetics and identify potential future tools for clinicians. In particular, P nuclear magnetic resonance to measure high-energy phosphates, serum lactate measurements, the use of near-infrared spectroscopy to measure the oxidation state of cytochrome aa3, and the ability of the protoporphyrin IX-triplet state lifetime technique to measure mitochondrial oxygen tension are discussed. In addition, this review discusses novel treatment strategies such as hyperbaric oxygen, preconditioning, exercise training, therapeutic gases, as well as inhibitors of HIF-1, HIF prolyl hydroxylase, and peroxisome proliferator-activated receptors.


Asunto(s)
Metabolismo Energético , Terapia por Ejercicio , Oxigenoterapia Hiperbárica , Oxígeno/metabolismo , Oxígeno/uso terapéutico , Sustancias Protectoras/uso terapéutico , Transducción de Señal , Adaptación Fisiológica , Animales , Biomarcadores/sangre , Monóxido de Carbono/uso terapéutico , Hipoxia de la Célula , Microambiente Celular , Metabolismo Energético/efectos de los fármacos , Inhibidores Enzimáticos/uso terapéutico , Gases , Humanos , Sulfuro de Hidrógeno/uso terapéutico , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Factor 1 Inducible por Hipoxia/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/antagonistas & inhibidores , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Espectroscopía de Resonancia Magnética/métodos , Óxido Nítrico/uso terapéutico , Receptores Activados del Proliferador del Peroxisoma/agonistas , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Sustancias Protectoras/metabolismo , Transducción de Señal/efectos de los fármacos , Espectroscopía Infrarroja Corta
17.
J Enzyme Inhib Med Chem ; 32(1): 992-1001, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28766956

RESUMEN

While progress has been made in treating cancer, cytotoxic chemotherapeutic agents are still the most widely used drugs and are associated with severe side-effects. Drugs that target unique molecular signalling pathways are needed for treating cancer with low or no intrinsic toxicity to normal cells. Our goal is to target hypoxic tumours and specifically the hypoxia inducible factor (HIF) pathway for the development of new cancer therapies. To this end, we have previously developed benzopyran-based HIF-1 inhibitors such as arylsulfonamide KCN1. However, KCN1 and its earlier analogs have poor water solubility, which hamper their applications. Herein, we describe a series of KCN1 analogs that incorporate a morpholine moiety at various positions. We found that replacing the benzopyran group of KCN1 with a phenyl group with a morpholinomethyl moiety at the para positions had minimal effect on potency and improved the water solubility of two new compounds by more than 10-fold compared to KCN1, the lead compound.


Asunto(s)
Benzopiranos/farmacología , Diseño de Fármacos , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Benzopiranos/síntesis química , Benzopiranos/química , Solubilidad , Agua/química
18.
Proc Natl Acad Sci U S A ; 111(50): E5429-38, 2014 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-25453096

RESUMEN

Triple negative breast cancers (TNBCs) are defined by the lack of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 expression, and are treated with cytotoxic chemotherapy such as paclitaxel or gemcitabine, with a durable response rate of less than 20%. TNBCs are enriched for the basal subtype gene expression profile and the presence of breast cancer stem cells, which are endowed with self-renewing and tumor-initiating properties and resistance to chemotherapy. Hypoxia-inducible factors (HIFs) and their target gene products are highly active in TNBCs. Here, we demonstrate that HIF expression and transcriptional activity are induced by treatment of MDA-MB-231, SUM-149, and SUM-159, which are human TNBC cell lines, as well as MCF-7, which is an ER(+)/PR(+) breast cancer line, with paclitaxel or gemcitabine. Chemotherapy-induced HIF activity enriched the breast cancer stem cell population through interleukin-6 and interleukin-8 signaling and increased expression of multidrug resistance 1. Coadministration of HIF inhibitors overcame the resistance of breast cancer stem cells to paclitaxel or gemcitabine, both in vitro and in vivo, leading to tumor eradication. Increased expression of HIF-1α or HIF target genes in breast cancer biopsies was associated with decreased overall survival, particularly in patients with basal subtype tumors and those treated with chemotherapy alone. Based on these results, clinical trials are warranted to test whether treatment of patients with TNBC with a combination of cytotoxic chemotherapy and HIF inhibitors will improve patient survival.


Asunto(s)
Resistencia a Antineoplásicos/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Análisis de Varianza , Línea Celular Tumoral , Cartilla de ADN/genética , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Humanos , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Procesamiento de Imagen Asistido por Computador , Immunoblotting , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Estimación de Kaplan-Meier , Luciferasas , Microscopía , Paclitaxel/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/metabolismo , Gemcitabina
19.
Cancer Sci ; 107(8): 1151-8, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27270607

RESUMEN

Pancreatic cancer is one of the most lethal digestive system cancers with a 5-year survival rate of 4-7%. Despite extensive efforts, recent chemotherapeutic regimens have provided only limited benefits to pancreatic cancer patients. Gemcitabine and TS-1, the current standard-of-care chemotherapeutic drugs for treatment of this severe cancer, have a low response rate. Hypoxia is one of the factors contributing to treatment resistance. Specifically, overexpression of hypoxia-inducible factor, a master transcriptional regulator of cell adaption to hypoxia, is strongly correlated with poor prognosis in many human cancers. TAT-ODD-procaspase-3 (TOP3) is a protein prodrug that is specifically processed and activated in hypoxia-inducible factor-active cells in cancers, leading to cell death. Here, we report combination therapies in which TOP3 was combined with gemcitabine or TS-1. As monotherapy, gemcitabine and TS-1 showed a limited effect on hypoxic and starved pancreatic cancer cells, whereas co-treatment with TOP3 successfully overcame this limitation in vitro. Furthermore, combination therapies of TOP3 with these drugs resulted in a significant improvement in survival of orthotopic pancreatic cancer models involving the human pancreatic cancer cell line SUIT-2. Overall, our study indicates that the combination of TOP3 with current chemotherapeutic drugs can significantly improve treatment outcome, offering a promising new therapeutic option for patients with pancreatic cancer.


Asunto(s)
Desoxicitidina/análogos & derivados , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Profármacos/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Silicatos/farmacología , Titanio/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Línea Celular Tumoral , Desoxicitidina/administración & dosificación , Desoxicitidina/farmacología , Fluorouracilo/farmacología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Profármacos/farmacología , Proteínas Recombinantes de Fusión/farmacología , Silicatos/administración & dosificación , Análisis de Supervivencia , Titanio/administración & dosificación , Hipoxia Tumoral , Gemcitabina
20.
Cancer Sci ; 107(6): 746-54, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27009878

RESUMEN

Cisplatin-resistant A549 and H157 (A549CisR and H157CisR) non-small cell lung cancer cells show increased stemness of cancer stem cells (CSCs) compared to their parental cells. We investigated whether interleukin-6 (IL-6) signaling contributes to this increased stemness in cisplatin-resistant cells. When A549CisR and H157CisR cells were treated with neutralizing IL-6 antibody, decreased cisplatin resistance was observed, whereas IL-6 treatment of parental cells resulted in increased cisplatin resistance. Expression of the CSC markers was significantly upregulated in IL-6-expressing scramble cells (in vitro) and scramble cell-derived tumor tissues (in vivo) after cisplatin treatment, but not in IL-6 knocked down (IL-6si) (in vitro) cells and in IL-6si cell-derived tumor tissues (in vivo), suggesting the importance of IL-6 signaling in triggering increased stemness during cisplatin resistance development. Hypoxia inducible factors (HIFs) were upregulated by IL-6 and responsible for the increased CSC stemness on cisplatin treatment. Mechanism dissection studies found that upregulation of HIFs by IL-6 was through transcriptional control and inhibition of HIF degradation. Treatment of HIF inhibitor (FM19G11) abolished the upregulation of CSC markers and increased sphere formations in IL-6 expressing cells on cisplatin treatment. In all, IL-6-mediated HIF upregulation is important in increasing stemness during cisplatin resistance development, and we suggest that the strategies of inhibiting IL-6 signaling or its downstream HIF molecules can be used as future therapeutic approaches to target CSCs after cisplatin treatment for lung cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Cisplatino/farmacología , Factor 1 Inducible por Hipoxia/biosíntesis , Interleucina-6/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Animales , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Cisplatino/uso terapéutico , Femenino , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Factor 1 Inducible por Hipoxia/genética , Factor 1 Inducible por Hipoxia/metabolismo , Interleucina-6/biosíntesis , Interleucina-6/genética , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Desnudos , Complejo de la Endopetidasa Proteasomal/metabolismo , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA