Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 80(6): 157, 2023 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-37208522

RESUMEN

Virilizer-like m6A methyltransferase-associated protein (VIRMA) maintains the stability of the m6A writer complex. Although VIRMA is critical for RNA m6A deposition, the impact of aberrant VIRMA expression in human diseases remains unclear. We show that VIRMA is amplified and overexpressed in 15-20% of breast cancers. Of the two known VIRMA isoforms, the nuclear-enriched full-length but not the cytoplasmic-localised N-terminal VIRMA promotes m6A-dependent breast tumourigenesis in vitro and in vivo. Mechanistically, we reveal that VIRMA overexpression upregulates the m6A-modified long non-coding RNA, NEAT1, which contributes to breast cancer cell growth. We also show that VIRMA overexpression enriches m6A on transcripts that regulate the unfolded protein response (UPR) pathway but does not promote their translation to activate the UPR under optimal growth conditions. Under stressful conditions that are often present in tumour microenvironments, VIRMA-overexpressing cells display enhanced UPR and increased susceptibility to death. Our study identifies oncogenic VIRMA overexpression as a vulnerability that may be exploited for cancer therapy.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Respuesta de Proteína Desplegada/genética , ARN/metabolismo , Interferencia de ARN , Microambiente Tumoral
2.
J Biol Chem ; 298(11): 102536, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36174675

RESUMEN

The cellular response to hypoxia is regulated through enzymatic oxygen sensors, including the prolyl hydroxylases, which control degradation of the well-known hypoxia inducible factors (HIFs). Other enzymatic oxygen sensors have been recently identified, including members of the KDM histone demethylase family. Little is known about how different oxygen-sensing pathways interact and if this varies depending on the form of hypoxia, such as chronic or intermittent. In this study, we investigated how two proposed cellular oxygen-sensing systems, HIF-1 and KDM4A, KDM4B, and KDM4C, respond in cells exposed to rapid forms of intermittent hypoxia (minutes) and compared to chronic hypoxia (hours). We found that intermittent hypoxia increases HIF-1α protein through a pathway distinct from chronic hypoxia, involving the KDM4A, KDM4B, and KDM4C histone lysine demethylases. Intermittent hypoxia increases the quantity and activity of KDM4A, KDM4B, and KDM4C, resulting in a decrease in histone 3 lysine 9 (H3K9) trimethylation near the HIF1A locus. We demonstrate that this contrasts with chronic hypoxia, which decreases KDM4A, KDM4B, and KDM4C activity, leading to hypertrimethylation of H3K9 globally and at the HIF1A locus. Altogether, we found that demethylation of histones bound to the HIF1A gene in intermittent hypoxia increases HIF1A mRNA expression, which has the downstream effect of increasing overall HIF-1 activity and expression of HIF target genes. This study highlights how multiple oxygen-sensing pathways can interact to regulate and fine tune the cellular hypoxic response depending on the period and length of hypoxia.


Asunto(s)
Histonas , Subunidad alfa del Factor 1 Inducible por Hipoxia , Procesamiento Proteico-Postraduccional , Humanos , Desmetilación , Histona Demetilasas/metabolismo , Histonas/genética , Histonas/metabolismo , Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Oxígeno/metabolismo
3.
Int J Mol Sci ; 22(14)2021 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-34298883

RESUMEN

As the cornerstone of high-grade glioma (HGG) treatment, radiotherapy temporarily controls tumor cells via inducing oxidative stress and subsequent DNA breaks. However, almost all HGGs recur within months. Therefore, it is important to understand the underlying mechanisms of radioresistance, so that novel strategies can be developed to improve the effectiveness of radiotherapy. While currently poorly understood, radioresistance appears to be predominantly driven by altered metabolism and hypoxia. Glucose is a central macronutrient, and its metabolism is rewired in HGG cells, increasing glycolytic flux to produce energy and essential metabolic intermediates, known as the Warburg effect. This altered metabolism in HGG cells not only supports cell proliferation and invasiveness, but it also contributes significantly to radioresistance. Several metabolic drugs have been used as a novel approach to improve the radiosensitivity of HGGs, including dichloroacetate (DCA), a small molecule used to treat children with congenital mitochondrial disorders. DCA reverses the Warburg effect by inhibiting pyruvate dehydrogenase kinases, which subsequently activates mitochondrial oxidative phosphorylation at the expense of glycolysis. This effect is thought to block the growth advantage of HGGs and improve the radiosensitivity of HGG cells. This review highlights the main features of altered glucose metabolism in HGG cells as a contributor to radioresistance and describes the mechanism of action of DCA. Furthermore, we will summarize recent advances in DCA's pre-clinical and clinical studies as a radiosensitizer and address how these scientific findings can be translated into clinical practice to improve the management of HGG patients.


Asunto(s)
Ácido Dicloroacético/farmacología , Glioma/tratamiento farmacológico , Glucosa/metabolismo , Tolerancia a Radiación/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Glioma/metabolismo , Glucólisis/efectos de los fármacos , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fosforilación Oxidativa/efectos de los fármacos
4.
Am J Physiol Cell Physiol ; 319(3): C533-C540, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32726159

RESUMEN

Humans have internal circadian clocks that ensure that important physiological functions occur at specific times of the day. These molecular clocks are regulated at the genomic level and exist in most cells of the body. Multiple circadian resetting cues have been identified, including light, temperature, and food. Recently, oxygen has been identified as a resetting cue, and emerging science indicates that this occurs through interactions at the cellular level between the circadian transcription-translation feedback loop and the hypoxia-inducible pathway (hypoxia-inducible factor; subject of the 2019 Nobel Prize in Physiology or Medicine). This review will cover recently identified relationships between HIF and proteins of the circadian clock. Interactions between the circadian clock and hypoxia could have wide-reaching implications for human diseases, and understanding the molecular mechanisms regulating these overlapping pathways may open up new strategies for drug discovery.


Asunto(s)
Relojes Circadianos/genética , Ritmo Circadiano/fisiología , Factor 1 Inducible por Hipoxia/metabolismo , Factores de Tiempo , Animales , Descubrimiento de Drogas , Humanos , Hipoxia/metabolismo
5.
Cell Physiol Biochem ; 53(3): 480-495, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31486323

RESUMEN

BACKGROUND/AIMS: Hypoxia Inducible Factor-1α (HIF-1α) is involved in cancer progression and is stabilized by the chaperone HSP90 (Heat Shock Protein 90), preventing degradation. Previously identified HSP90 inhibitors bind to the N-terminal pocket of HSP90, which blocks binding to HIF-1α and induces HIF-1α degradation. N-terminal inhibitors have failed in the clinic as single therapy treatments partially because they induce a heat shock response. SM molecules are HSP90 inhibitors that bind to the C-terminus of HSP90 and do not induce a heat shock response. The effects of these C-terminal inhibitors on HIF-1α are unreported. METHODS: HCT116, MDA-MB-231, PC3, and HEK293T cells were treated with HSP90 inhibitors. qRT-PCR and western blotting was performed to assess mRNA and protein levels of HIF-1α, HSP- and RACK1-related genes. siRNA was used to knockdown RACK1, while MG262 was used to inhibit proteasome activity. Dimethyloxalylglycine (DMOG) was used to inhibit activity of the prolyl hydroxylases (PHDs). Anti-angiogenic activity of HSP90 inhibitors was assessed using a HUVEC tubule formation assay. RESULTS: We show that SM compounds decrease HIF-1α target expression at the mRNA and protein level under hypoxia in colorectal, breast and prostate cancer cells, leading to cell death, without inducing a heat shock response. Surprisingly, we found that when the C-terminal of HSP90 is inhibited, HIF-1α degradation occurs through the proteasome and prolyl hydroxylases in an oxygen-dependent manner even in very low levels of oxygen (tumor hypoxia levels). RACK1 was not required for proteasomal degradation of HIF-1α. CONCLUSION: Our results suggest that by targeting the C-terminus of HSP90 we can exploit the prolyl hydroxylase and proteasome pathway to induce HIF-1α degradation in hypoxic tumors.


Asunto(s)
Hipoxia de la Célula/fisiología , Proteínas HSP90 de Choque Térmico/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Aminoácidos Dicarboxílicos/metabolismo , Western Blotting , Hipoxia de la Célula/genética , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Células HCT116 , Células HEK293 , Proteínas HSP90 de Choque Térmico/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Células PC-3 , Prolil Hidroxilasas/genética , Prolil Hidroxilasas/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
7.
Int J Mol Sci ; 20(2)2019 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-30669593

RESUMEN

Obstructive sleep apnea (OSA) affects a significant proportion of the population and is linked to increased rates of cancer development and a worse cancer outcome. OSA is characterized by nocturnal intermittent hypoxia and animal models of OSA-like intermittent hypoxia show increased tumor growth and metastasis. Advanced tumors typically have regions of chronic hypoxia, activating the transcription factor, HIF-1, which controls the expression of genes involved in cancer progression. Rapid intermittent hypoxia from OSA has been proposed to increase HIF-1 activity and this may occur in tumors. The effect of exposing a developing tumor to OSA-like intermittent hypoxia is largely unknown. We have built a cell-based model of physiological OSA tissue oxygenation in order to study the effects of intermittent hypoxia in HCT116 colorectal cancer cells. We found that HIF-1α increases following intermittent hypoxia and that the expression of HIF-target genes increases, including those involved in glycolysis, the hypoxic pathway and extracellular matrix remodeling. Expression of these genes acts as a 'hypoxic' signature which is associated with a worse prognosis. The total dose of hypoxia determined the magnitude of change in the hypoxic signature rather than the frequency or duration of hypoxia-reoxygenation cycles per se. Finally, transcription of HIF1A mRNA differs in response to chronic and intermittent hypoxia suggesting that HIF-1α may be regulated at the transcriptional level in intermittent hypoxia and not just by the post-translational oxygen-dependent degradation pathway seen in chronic hypoxia.


Asunto(s)
Factor 1 Inducible por Hipoxia/genética , Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/genética , Hipoxia/metabolismo , Apnea Obstructiva del Sueño/genética , Apnea Obstructiva del Sueño/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Matriz Extracelular , Regulación de la Expresión Génica , Glucólisis , Células HCT116 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Modelos Biológicos , Interferencia de ARN , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Transducción de Señal
8.
Am J Physiol Regul Integr Comp Physiol ; 315(4): R669-R687, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29995459

RESUMEN

Obstructive sleep apnea (OSA) is common and linked to a variety of poor health outcomes. A key modulator of this disease is nocturnal intermittent hypoxia. There is striking epidemiological evidence that patients with OSA have higher rates of cancer and cancer mortality. Small-animal models demonstrate an important role for systemic intermittent hypoxia in tumor growth and metastasis, yet the underlying mechanisms are poorly understood. Emerging data indicate that intermittent hypoxia activates the hypoxic response and inflammatory pathways in a manner distinct from chronic hypoxia. However, there is significant heterogeneity in published methods for modeling hypoxic conditions, which are often lacking in physiological relevance. This is particularly important for studying key transcriptional mediators of the hypoxic and inflammatory responses such as hypoxia-inducible factor (HIF) and NF-κB. The relationship between HIF, the molecular clock, and circadian rhythm may also contribute to cancer risk in OSA. Building accurate in vitro models of intermittent hypoxia reflective of OSA is challenging but necessary to better elucidate underlying molecular pathways.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Hipoxia/metabolismo , Neoplasias/metabolismo , Oxígeno/metabolismo , Apnea Obstructiva del Sueño/metabolismo , Animales , Hipoxia de la Célula , Movimiento Celular , Proliferación Celular , Transformación Celular Neoplásica/patología , Péptidos y Proteínas de Señalización del Ritmo Circadiano/metabolismo , Modelos Animales de Enfermedad , Humanos , Hipoxia/epidemiología , Incidencia , Mediadores de Inflamación/metabolismo , Metástasis de la Neoplasia , Neoplasias/epidemiología , Neoplasias/patología , Factores de Riesgo , Transducción de Señal , Apnea Obstructiva del Sueño/epidemiología , Microambiente Tumoral
9.
Mar Drugs ; 16(7)2018 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-30029505

RESUMEN

Elements of the hypoxia inducible factor (HIF) transcriptional system, a key regulator of the cellular hypoxic response, are up-regulated in a range of cancer cells. HIF is fundamentally involved in tumor angiogenesis, invasion, and energy metabolism. Inhibition of the transcriptional activity of HIF may be of therapeutic benefit to cancer patients. We recently described the identification of two marine pyrroloiminoquinone alkaloids with potent activity in inhibiting the interaction between the oncogenic transcription factor HIF-1α and the coactivator protein p300. Herein, we present further characterization data for these two screening hits: discorhabdin H (1) and discorhabdin L (2), with a specific focus on their anti-angiogenic and anti-tumor effects. We demonstrated that only discorhabdin L (2) possesses excellent anti-angiogenic activity in inhibiting endothelial cell proliferation and tube formation, as well as decreasing microvessel outgrowth in the ex vivo rat aortic ring assay. We further showed that discorhabdin L (2) significantly inhibits in vivo prostate tumor growth in a LNCaP xenograft model. In conclusion, our findings suggest that discorhabdin L (2) represents a promising HIF-1α inhibitor worthy of further drug development.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacocinética , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Neovascularización Patológica/tratamiento farmacológico , Quinonas/farmacología , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Proteína p300 Asociada a E1A/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Ratones , Ratones SCID , Neovascularización Patológica/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos
10.
CA Cancer J Clin ; 60(4): 222-43, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20554717

RESUMEN

Angiogenesis has become an attractive target for drug therapy because of its key role in tumor growth. An extensive array of compounds is currently in preclinical development, with many now entering the clinic and/or achieving approval from the US Food and Drug Administration. Several regulatory and signaling molecules governing angiogenesis are of interest, including growth factors (eg, vascular endothelial growth factor, platelet-derived growth factor, fibroblast growth factor, and epidermal growth factor), receptor tyrosine kinases, and transcription factors such as hypoxia inducible factor, as well as molecules involved in mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) signaling. Pharmacologic agents have been identified that target these pathways, yet for some agents (notably thalidomide), an understanding of the specific mechanisms of antitumor action has proved elusive. The following review describes key molecular mechanisms and novel therapies that are on the horizon for antiangiogenic tumor therapy.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Neoplasias/tratamiento farmacológico , Inhibidores de la Angiogénesis/farmacología , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Transformación Celular Neoplásica , Farnesiltransferasa/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/fisiología , Neoplasias/irrigación sanguínea , Neoplasias/metabolismo , Neovascularización Patológica/prevención & control , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Inhibidores de la Síntesis del Ácido Nucleico/uso terapéutico , Unión Proteica/efectos de los fármacos , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores Notch/antagonistas & inhibidores , Receptores Notch/fisiología , Transducción de Señal/efectos de los fármacos , Tiorredoxinas/antagonistas & inhibidores
11.
Blood ; 123(13): 2000-7, 2014 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-24523239

RESUMEN

Most proteins in nature are chemically modified after they are made to control how, when, and where they function. The 3 core features of proteins are posttranslationally modified: amino acid side chains can be modified, peptide bonds can be cleaved or isomerized, and disulfide bonds can be cleaved. Cleavage of peptide bonds is a major mechanism of protein control in the circulation, as exemplified by activation of the blood coagulation and complement zymogens. Cleavage of disulfide bonds is emerging as another important mechanism of protein control in the circulation. Recent advances in our understanding of control of soluble blood proteins and blood cell receptors by functional disulfide bonds is discussed as is how these bonds are being identified and studied.


Asunto(s)
Regulación Alostérica/fisiología , Proteínas Sanguíneas/química , Proteínas Sanguíneas/metabolismo , Disulfuros/química , Angiotensinógeno/química , Angiotensinógeno/metabolismo , Animales , Disulfuros/metabolismo , Humanos , Enlace de Hidrógeno , Subunidad gamma Común de Receptores de Interleucina/química , Subunidad gamma Común de Receptores de Interleucina/metabolismo , Plasminógeno/química , Plasminógeno/metabolismo , beta 2 Glicoproteína I/química , beta 2 Glicoproteína I/metabolismo
12.
J Nat Prod ; 79(5): 1267-75, 2016 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-27140429

RESUMEN

Inhibition of the hypoxia-inducible factor 1α (HIF-1α) pathway by disrupting its association with the transcriptional coactivator p300 inhibits angiogenesis and tumor development. Development of HIF-1α/p300 inhibitors has been hampered by preclinical toxicity; therefore, we aimed to identify novel HIF-1α/p300 inhibitors. Using a cell-free assay designed to test compounds that block HIF-1α/p300 binding, 170 298 crude natural product extracts and prefractionated samples were screened, identifying 25 active extracts. One of these extracts, originating from the marine sponge Latrunculia sp., afforded six pyrroloiminoquinone alkaloids that were identified as positive hits (IC50 values: 1-35 µM). Luciferase assays confirmed inhibition of HIF-1α transcriptional activity by discorhabdin B (1) and its dimer (2), 3-dihydrodiscorhabdin C (3), makaluvamine F (5), discorhabdin H (8), discorhabdin L (9), and discorhabdin W (11) in HCT 116 colon cancer cells (0.1-10 µM, p < 0.05). Except for 11, all of these compounds also reduced HIF-1α transcriptional activity in LNCaP prostate cancer cells (0.1-10 µM, p < 0.05). These effects occurred at noncytotoxic concentrations (<50% cell death) under hypoxic conditions. At the downstream HIF-1α target level, compound 8 (0.5 µM) significantly decreased VEGF secretion in LNCaP cells (p < 0.05). In COLO 205 colon cancer cells no activity was shown in the luciferase or cytotoxicity assays. Pyrroloiminoquinone alkaloids are a novel class of HIF-1α inhibitors, which interrupt the protein-protein interaction between HIF-1α and p300 and consequently reduce HIF-related transcription.


Asunto(s)
Alcaloides/farmacología , Proteína p300 Asociada a E1A/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Poríferos/química , Pirroliminoquinonas/farmacología , Alcaloides/química , Animales , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células HCT116 , Compuestos Heterocíclicos de 4 o más Anillos , Humanos , Masculino , Biología Marina , Estructura Molecular , Neovascularización Patológica , Neoplasias de la Próstata/tratamiento farmacológico , Pirroliminoquinonas/química , Quinonas , Compuestos de Espiro , Tiazepinas , Factor A de Crecimiento Endotelial Vascular/metabolismo
13.
J Am Chem Soc ; 137(16): 5569-75, 2015 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-25892103

RESUMEN

Low oxygen environments are a hallmark of solid tumors, and transcription of many hypoxia-responsive genes needed for survival under these conditions is regulated by the transcription factor HIF-1 (hypoxia-inducible factor 1). Activation of HIF-1 requires binding of its α-subunit (HIF-1α) to the transcriptional coactivator protein p300. Inhibition of the p300/HIF-1α interaction can suppress HIF-1 activity. A screen for inhibitors of the protein binding domains of p300 (CH1) and HIF-1α (C-TAD) identified an extract of the marine ascidian Eudistoma sp. as active. Novel heterocyclic alkaloids eudistidines A (1) and B (2) were isolated from the extract, and their structures assigned by spectroscopic analyses. They contain an unprecedented tetracyclic core composed of two pyrimidine rings fused with an imidazole ring. Eudistidine A (1) was synthesized in a concise four-step sequence featuring a condensation/cyclization reaction cascade between 4-(2-aminophenyl)pyrimidin-2-amine (3) and 4-methoxy-phenylglyoxal (4), while eudistidine B (2) was synthesized in a similar fashion with glyoxylic acid (5) in place of 4. Naturally occurring eudistidine A (1) effectively inhibited CH1/C-TAD binding with an IC50 of 75 µM, and synthetic 1 had similar activity. The eudistidine A (1) scaffold, which can be synthesized in a concise, scalable manner, may provide potential therapeutic lead compounds or molecular probes to study p300/HIF-1α interactions and the role these proteins play in tumor response to low oxygen conditions. The unique structural scaffolds and functional group arrays often found in natural products make these secondary metabolites a rich source of new compounds that can disrupt critical protein-protein binding events.


Asunto(s)
Alcaloides/química , Alcaloides/farmacología , Proteína p300 Asociada a E1A/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Compuestos Policíclicos/química , Compuestos Policíclicos/farmacología , Mapas de Interacción de Proteínas/efectos de los fármacos , Alcaloides/síntesis química , Animales , Proteína p300 Asociada a E1A/química , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/química , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Compuestos Policíclicos/síntesis química , Unión Proteica/efectos de los fármacos , Urocordados/química
14.
J Biol Chem ; 288(48): 34920-9, 2013 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-24142694

RESUMEN

The S1A serine proteases function in many key biological processes such as development, immunity, and blood coagulation. S1A proteases contain a highly conserved disulfide bond (Cys(191)-Cys(220) in chymotrypsin numbering) that links two ß-loop structures that define the rim of the active site pocket. Mast cell ßII-tryptase is a S1A protease that is associated with pathological inflammation. In this study, we have found that the conserved disulfide bond (Cys(220)-Cys(248) in ßII-tryptase) exists in oxidized and reduced states in the enzyme stored and secreted by mast cells. The disulfide bond has a standard redox potential of -301 mV and is stoichiometrically reduced by the inflammatory mediator, thioredoxin, with a rate constant of 350 m(-1) s(-1). The oxidized and reduced enzymes have different substrate specificity and catalytic efficiency for hydrolysis of both small and macromolecular substrates. These observations indicate that ßII-tryptase activity is post-translationally regulated by an allosteric disulfide bond. It is likely that other S1A serine proteases are similarly regulated.


Asunto(s)
Cisteína/química , Mastocitos/enzimología , Oxidación-Reducción , Triptasas/química , Regulación Alostérica , Animales , Sitios de Unión , Catálisis , Dominio Catalítico , Línea Celular Tumoral , Cisteína/metabolismo , Disulfuros/química , Disulfuros/metabolismo , Humanos , Mastocitos/metabolismo , Ratones , Relación Estructura-Actividad , Especificidad por Sustrato , Triptasas/metabolismo
15.
Mol Cancer ; 13: 91, 2014 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-24775564

RESUMEN

The downstream targets of hypoxia inducible factor-1 alpha (HIF-1α) play an important role in tumor progression and angiogenesis. Therefore, inhibition of HIF-mediated transcription has potential in the treatment of cancer. One attractive strategy for inhibiting HIF activity is the disruption of the HIF-1α/p300 complex, as p300 is a crucial coactivator of hypoxia-inducible transcription. Several members of the epidithiodiketopiperazine (ETP) family of natural products have been shown to disrupt the HIF-1α/p300 complex in vitro; namely, gliotoxin, chaetocin, and chetomin. Here, we further characterized the molecular mechanisms underlying the antiangiogenic and antitumor effects of these ETPs using a preclinical model of prostate cancer. In the rat aortic ring angiogenesis assay, gliotoxin, chaetocin, and chetomin significantly inhibited microvessel outgrowth at a GI50 of 151, 8, and 20 nM, respectively. In vitro co-immunoprecipitation studies in prostate cancer cell extracts demonstrated that these compounds disrupted the HIF-1α/p300 complex. The downstream effects of inhibiting the HIF-1α/p300 interaction were evaluated by determining HIF-1α target gene expression at the mRNA and protein levels. Dose-dependent decreases in levels of secreted VEGF were detected by ELISA in the culture media of treated cells, and the subsequent downregulation of VEGFA, LDHA, and ENO1 HIF-1α target genes were confirmed by semi-quantitative real-time PCR. Finally, treatment with ETPs in mice bearing prostate tumor xenografts resulted in significant inhibition of tumor growth. These results suggest that directly targeting the HIF-1α/p300 complex with ETPs may be an effective approach for inhibiting angiogenesis and tumor growth.


Asunto(s)
Antineoplásicos/farmacología , Proteína p300 Asociada a E1A/genética , Regulación Neoplásica de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Disulfuros/farmacología , Proteína p300 Asociada a E1A/antagonistas & inhibidores , Proteína p300 Asociada a E1A/metabolismo , Células Endoteliales/efectos de los fármacos , Gliotoxina/farmacología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Alcaloides Indólicos/farmacología , Isoenzimas/genética , Isoenzimas/metabolismo , L-Lactato Deshidrogenasa/genética , L-Lactato Deshidrogenasa/metabolismo , Lactato Deshidrogenasa 5 , Masculino , Trasplante de Neoplasias , Neovascularización Patológica/prevención & control , Fosfopiruvato Hidratasa/genética , Fosfopiruvato Hidratasa/metabolismo , Piperazinas/farmacología , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/patología , Unión Proteica/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Transducción de Señal , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
J Med Imaging Radiat Oncol ; 66(4): 560-574, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35466515

RESUMEN

Immune checkpoint inhibition with PD-1/PD-L1 blockade is a promising area in the field of anti-cancer therapy. Although clinical data have revealed success of PD-1/PD-L1 blockade as monotherapy or in combination with CTLA-4 or chemotherapy, the combination with radiotherapy could further boost anti-tumour immunity and enhance clinical outcomes due to the immunostimulatory effects of radiation. However, the synergistic combination of PD-1/PD-L1 blockade and radiotherapy can be challenged by the complex nature of the tumour microenvironment (TME), including the presence of tumour hypoxia. Hypoxia is a major barrier to the effectiveness of both radiotherapy and PD-1/PD-L1 blockade immunotherapy. Thus, targeting the hypoxic TME is an attractive strategy to enhance the efficacy of the combination. Addition of compounds that directly or indirectly reduce hypoxia, to the combination of PD-1/PD-L1 inhibitors and radiotherapy may optimize the success of the combination and improve therapeutic outcomes. In this review, we will discuss the synergistic combination of PD-1/PD-L1 blockade and radiotherapy and highlight the role of hypoxic TME in impeding the success of both therapies. In addition, we will address the potential approaches for targeting tumour hypoxia and how exploiting these strategies could benefit the combination of PD-1/PD-L1 blockade and radiotherapy.


Asunto(s)
Antígeno B7-H1 , Neoplasias , Antígeno B7-H1/farmacología , Antígeno B7-H1/uso terapéutico , Humanos , Hipoxia/tratamiento farmacológico , Inhibidores de Puntos de Control Inmunológico , Neoplasias/radioterapia , Receptor de Muerte Celular Programada 1/uso terapéutico , Microambiente Tumoral
17.
J Biol Chem ; 284(39): 26831-8, 2009 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-19589782

RESUMEN

The hypoxic response in humans is regulated by the hypoxia-inducible transcription factor system; inhibition of hypoxia-inducible factor (HIF) activity has potential for the treatment of cancer. Chetomin, a member of the epidithiodiketopiperazine (ETP) family of natural products, inhibits the interaction between HIF-alpha and the transcriptional coactivator p300. Structure-activity studies employing both natural and synthetic ETP derivatives reveal that only the structurally unique ETP core is required and sufficient to block the interaction of HIF-1alpha and p300. In support of both cell-based and animal work showing that the cytotoxic effect of ETPs is reduced by the addition of Zn(2+) through an unknown mechanism, our mechanistic studies reveal that ETPs react with p300, causing zinc ion ejection. Cell studies with both natural and synthetic ETPs demonstrated a decrease in vascular endothelial growth factor and antiproliferative effects that were abrogated by zinc supplementation. The results have implications for the design of selective ETPs and for the interaction of ETPs with other zinc ion-binding protein targets involved in gene expression.


Asunto(s)
Disulfuros/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Alcaloides Indólicos/farmacología , Zinc/farmacología , Factores de Transcripción p300-CBP/metabolismo , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Disulfuros/química , Relación Dosis-Respuesta a Droga , Células HCT116 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/química , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Alcaloides Indólicos/química , Modelos Químicos , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Espectrometría de Masa por Ionización de Electrospray , Relación Estructura-Actividad , Factor A de Crecimiento Endotelial Vascular/metabolismo , Zinc/química , Factores de Transcripción p300-CBP/química , Factores de Transcripción p300-CBP/genética
18.
FEBS J ; 287(18): 3917-3920, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32652856

RESUMEN

Comorbidities are an important factor in tuberculosis pathophysiology and treatment but are understudied in animal models. Schild et al. present a zebrafish model of Mycobacterium marinum infection and wound comorbidity that retains responsiveness to protective hypoxia-inducible factor-1α activation as an example of a host-directed therapy. This platform is a new paradigm for the zebrafish-M. marinum infection model and provides a blueprint to test therapeutic interventions on infection and comorbid pathologies. Comment on: https://doi.org/10.1111/febs.15433.


Asunto(s)
Infecciones por Mycobacterium no Tuberculosas , Mycobacterium marinum , Tuberculosis , Animales , Comorbilidad , Modelos Animales de Enfermedad , Subunidad alfa del Factor 1 Inducible por Hipoxia , Tuberculosis/tratamiento farmacológico , Tuberculosis/epidemiología , Tuberculosis/prevención & control , Pez Cebra
19.
Methods Mol Biol ; 1967: 65-86, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31069765

RESUMEN

The redox potential of a protein disulphide bond is one of the most important factors for determining the role of a disulphide bond. Disulphide bonds can have a stabilizing role for the structure of a protein or they can play a functional role which can regulate protein bioactivity. Determining the redox potential of disulphides can help distinguish the functional from the structural disulphide bonds. In this chapter, two methods for determining the redox potential of a protein disulphide bond are described. The first method uses maleimide-biotin labeling of free cysteine thiols and western blot densitometry to determine the fraction of reduced disulphide bond under various redox-buffering conditions. The second method uses differential cysteine labeling and tandem mass spectrometry to determine the redox potential.


Asunto(s)
Cisteína/química , Disulfuros/química , Proteínas/química , Espectrometría de Masas en Tándem/métodos , Biotina/química , Maleimidas/química , Oxidación-Reducción , Dominios Proteicos , Pliegue de Proteína , Proteínas/genética , Compuestos de Sulfhidrilo/química
20.
Bio Protoc ; 9(18): e3371, 2019 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-33654867

RESUMEN

Dissolved oxygen and its availability to cells in culture is an overlooked variable which can have significant consequences on experimental research outcomes, including reproducibility. Oxygen sensing pathways play key roles in cell growth and behavior and pericellular oxygen levels should be controlled when establishing in vitro models. Standard cell culture techniques do not have adequate control over pericellular oxygen levels. Slow diffusion through culture media limits the precision of oxygen delivery to cells, making it difficult to accurately reproduce in vivo-like oxygen conditions. Furthermore, different types of cells consume oxygen at varying rates and this can be affected by the density of growing cells. Here, we describe a novel in vitro system that utilizes hypoxic chambers and oxygen-permeable culture dishes to control pericellular oxygen levels and provide rapid oxygen delivery to adherent cells. This procedure is particularly relevant for protocols studying effects of rapid oxygen changes or intermittent hypoxia on cellular behavior. The system is inexpensive and easily assembled without highly specialized equipment.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA