Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cancers (Basel) ; 16(7)2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38611076

RESUMEN

Cancer cells undergo a significant level of "metabolic reprogramming" or "remodeling" to ensure an adequate supply of ATP and "building blocks" for cell survival and to facilitate accelerated proliferation. Cancer cells preferentially use glycolysis for ATP production (the Warburg effect); however, cancer cells, including colorectal cancer (CRC) cells, also depend on oxidative phosphorylation (OXPHOS) for ATP production, a finding that suggests that both glycolysis and OXPHOS play significant roles in facilitating cancer progression and proliferation. Our prior studies identified a semisynthetic isoflavonoid, DBI-1, that served as an AMPK activator targeting mitochondrial complex I. Furthermore, DBI-1 and a glucose transporter 1 (GLUT1) inhibitor, BAY-876, synergistically inhibited CRC cell growth in vitro and in vivo. We now report a study of the structure-activity relationships (SARs) in the isoflavonoid family in which we identified a new DBI-1 analog, namely, DBI-2, with promising properties. Here, we aimed to explore the antitumor mechanisms of DBIs and to develop new combination strategies by targeting both glycolysis and OXPHOS. We identified DBI-2 as a novel AMPK activator using an AMPK phosphorylation assay as a readout. DBI-2 inhibited mitochondrial complex I in the Seahorse assays. We performed proliferation and Western blotting assays and conducted studies of apoptosis, necrosis, and autophagy to corroborate the synergistic effects of DBI-2 and BAY-876 on CRC cells in vitro. We hypothesized that restricting the carbohydrate uptake with a KD would mimic the effects of GLUT1 inhibitors, and we found that a ketogenic diet significantly enhanced the therapeutic efficacy of DBI-2 in CRC xenograft mouse models, an outcome that suggested a potentially new approach for combination cancer therapy.

2.
Am J Cancer Res ; 13(3): 976-991, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37034206

RESUMEN

Lung cancer is the leading cause of cancer-related deaths. Lung cancer cells develop resistance to apoptosis by suppressing the secretion of the tumor suppressor Par-4 protein (also known as PAWR) and/or down-modulating the Par-4 receptor GRP78 on the cell surface (csGRP78). We sought to identify FDA-approved drugs that elevate csGRP78 on the surface of lung cancer cells and induce Par-4 secretion from the cancer cells and/or normal cells in order to inhibit cancer growth in an autocrine or paracrine manner. In an unbiased screen, we identified crizotinib (CZT), an inhibitor of activated ALK/MET/ROS1 receptor tyrosine kinase, as an inducer of csGRP78 expression in ALK-negative, KRAS or EGFR mutant lung cancer cells. Elevation of csGRP78 in the lung cancer cells was dependent on activation of the non-receptor tyrosine kinase SRC by CZT. Inhibition of SRC activation in the cancer cells prevented csGRP78 translocation but promoted Par-4 secretion by CZT, implying that activated SRC prevented Par-4 secretion. In normal cells, CZT did not activate SRC and csGRP78 elevation but induced Par-4 secretion. Consequently, CZT induced Par-4 secretion from normal cells and elevated csGRP78 in the ALK-negative tumor cells to cause paracrine apoptosis in cancer cell cultures and growth inhibition of tumor xenografts in mice. Thus, CZT induces differential activation of SRC in normal and cancer cells to trigger the pro-apoptotic Par-4-GRP78 axis. As csGRP78 is a targetable receptor, CZT can be repurposed to elevate csGRP78 for inhibition of ALK-negative lung tumors.

3.
Mol Cancer Ther ; 21(5): 740-750, 2022 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-35247917

RESUMEN

Cancer cells undergo significant "metabolic remodeling" to provide sufficient ATP to maintain cell survival and to promote rapid growth. In colorectal cancer cells, ATP is produced by mitochondrial oxidative phosphorylation and by substantially elevated cytoplasmic glucose fermentation (i.e., the Warburg effect). Glucose transporter 1 (GLUT1) expression is significantly increased in colorectal cancer cells, and GLUT1 inhibitors block glucose uptake and hence glycolysis crucial for cancer cell growth. In addition to ATP, these metabolic pathways also provide macromolecule building blocks and signaling molecules required for tumor growth. In this study, we identify a diaminobutoxy-substituted isoflavonoid (DBI-1) that inhibits mitochondrial complex I and deprives rapidly growing cancer cells of energy needed for growth. DBI-1 and the GLUT1 inhibitor, BAY-876, synergistically inhibit colorectal cancer cell growth in vitro and in vivo. This study suggests that an electron transport chain inhibitor (i.e., DBI-1) and a glucose transport inhibitor, (i.e., BAY-876) are potentially effective combination for colorectal cancer treatment.


Asunto(s)
Neoplasias del Colon , Humanos , Adenosina Trifosfato , Línea Celular Tumoral , Transformación Celular Neoplásica , Neoplasias del Colon/tratamiento farmacológico , Glucosa , Transportador de Glucosa de Tipo 1/genética , Glucólisis , Pirazoles , Quinolinas , Factores de Transcripción
4.
Neurobiol Aging ; 54: 163-169, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28385551

RESUMEN

Beta-amyloid (Aß) deposition in brain accumulates as a function of age in people with Down syndrome (DS) with subsequent development into Alzheimer disease neuropathology, typically by 40 years of age. In vivo imaging using the Pittsburgh compound B (PiB) ligand has facilitated studies linking Aß, cognition, and dementia in DS. However, there are no studies of PiB binding across the lifespan in DS. The current study describes in vitro 3H-PiB binding in the frontal cortex of autopsy cases with DS compared to non-DS controls. Tissue from 64 cases included controls (n = 25) and DS (n = 39). In DS, 3H-PiB binding was significantly associated with age. After age 40 years in DS, 3H-PiB binding rose dramatically along with increasing individual variability. 3H-PiB binding correlated with the amount of Aß42. Using fixed frontal tissue and fluorescent 6-CN-PiB, neuritic and cored plaques along with extensive cerebral amyloid angiopathy showed 6-CN-PiB binding. These results suggest that cortical PiB binding as shown by positron emission tomography imaging reflects plaques and cerebral amyloid angiopathy in DS brain.


Asunto(s)
Envejecimiento/metabolismo , Péptidos beta-Amiloides/metabolismo , Compuestos de Anilina/metabolismo , Síndrome de Down/metabolismo , Lóbulo Frontal/metabolismo , Tiazoles/metabolismo , Adolescente , Adulto , Anciano , Autopsia , Angiopatía Amiloide Cerebral/diagnóstico por imagen , Niño , Preescolar , Cognición , Síndrome de Down/diagnóstico por imagen , Síndrome de Down/psicología , Femenino , Lóbulo Frontal/diagnóstico por imagen , Humanos , Lactante , Ligandos , Masculino , Persona de Mediana Edad , Tomografía de Emisión de Positrones , Unión Proteica , Adulto Joven
5.
Nat Chem Biol ; 13(4): 366-368, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28166207

RESUMEN

This study highlights the biochemical and structural characterization of the L-tryptophan C6 C-prenyltransferase (C-PT) PriB from Streptomyces sp. RM-5-8. PriB was found to be uniquely permissive to a diverse array of prenyl donors and acceptors including daptomycin. Two additional PTs also produced novel prenylated daptomycins with improved antibacterial activities over the parent drug.


Asunto(s)
Dimetilaliltranstransferasa/química , Dimetilaliltranstransferasa/metabolismo , Streptomyces/enzimología , Modelos Moleculares , Estructura Molecular , Especificidad por Sustrato
6.
ACS Chem Biol ; 10(7): 1729-36, 2015 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-25897591

RESUMEN

As part of an effort to identify substrate analogs suitable for helping to resolve structural features important for terpene synthases, the inhibition of 5-epi-aristolochene biosynthesis from farnesyl diphosphate (FPP) by the tobacco 5-epi-aristolochene synthase incubated with anilinogeranyl diphosphate (AGPP) was examined. The apparent noncompetitive nature of the inhibition supported further assessment of how AGPP might be bound to crystallographic forms of the enzyme. Surprisingly, the bound form of the inhibitor appeared to have undergone a cyclization event consistent with the native mechanism associated with FPP catalysis. Biocatalytic formation of a novel 13-membered macrocyclic paracyclophane alkaloid was confirmed by high-resolution GC-MS and NMR analysis. This work provides insights into new biosynthetic means for generating novel, functionally diversified, medium-sized terpene alkaloids.


Asunto(s)
Alcaloides/metabolismo , Transferasas Alquil y Aril/metabolismo , Compuestos Macrocíclicos/metabolismo , Nicotiana/enzimología , Fosfatos de Poliisoprenilo/metabolismo , Sesquiterpenos/metabolismo , Alcaloides/química , Transferasas Alquil y Aril/química , Compuestos Macrocíclicos/química , Modelos Moleculares , Fosfatos de Poliisoprenilo/química , Sesquiterpenos/química , Terpenos/química , Terpenos/metabolismo , Nicotiana/química , Nicotiana/metabolismo
7.
Mol Biol Cell ; 26(10): 1845-56, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25808490

RESUMEN

The clinical interest in farnesyltransferase inhibitors (FTIs) makes it important to understand how these compounds affect cellular processes involving farnesylated proteins. Mitotic abnormalities observed after treatment with FTIs have so far been attributed to defects in the farnesylation of the outer kinetochore proteins CENP-E and CENP-F, which are involved in chromosome congression and spindle assembly checkpoint signaling. Here we identify the cytoplasmic dynein adaptor Spindly as an additional component of the outer kinetochore that is modified by farnesyltransferase (FTase). We show that farnesylation of Spindly is essential for its localization, and thus for the proper localization of dynein and its cofactor dynactin, to prometaphase kinetochores and that Spindly kinetochore recruitment is more severely affected by FTase inhibition than kinetochore recruitment of CENP-E and CENP-F. Molecular replacement experiments show that both Spindly and CENP-E farnesylation are required for efficient chromosome congression. The identification of Spindly as a new mitotic substrate of FTase provides insight into the causes of the mitotic phenotypes observed with FTase inhibitors.


Asunto(s)
Proteínas Portadoras/metabolismo , Inhibidores Enzimáticos/farmacología , Farnesiltransferasa/antagonistas & inhibidores , Cinetocoros/efectos de los fármacos , Mitosis/efectos de los fármacos , Animales , Proteínas Portadoras/antagonistas & inhibidores , Proteínas de Ciclo Celular , Proteínas Cromosómicas no Histona/metabolismo , Segregación Cromosómica/efectos de los fármacos , Complejo Dinactina , Dineínas/efectos de los fármacos , Farnesiltransferasa/metabolismo , Humanos , Cinetocoros/metabolismo , Proteínas Asociadas a Microtúbulos/efectos de los fármacos , Prenilación de Proteína , Xenopus
8.
Curr Chem Biol ; 9(2): 123-141, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-27099830

RESUMEN

BACKGROUND: Dolichyl phosphate-linked mono- and oligosaccharides (DLO) are essential intermediates in protein N-glycosylation, C- and O-mannosylation and GPI anchor biosynthesis. While many membrane proteins in the endoplasmic reticulum (ER) involved in the assembly of DLOs are known, essential proteins believed to be required for the transbilayer movement (flip-flopping) and proteins potentially involved in the regulation of DLO synthesis remain to be identified. METHODS: The synthesis of a series of Dol-P derivatives composed of citronellyl-based photoprobes with benzophenone groups equipped with alkyne moieties for Huisgen "click" chemistry is now described to utilize as tools for identifying ER proteins involved in regulating the biosynthesis and transbilayer movement of lipid intermediates. In vitro enzymatic assays were used to establish that the photoprobes contain the critical structural features recognized by pertinent enzymes in the dolichol pathway. ER proteins that photoreacted with the novel probes were identified by MS. RESULTS: The potential of the newly designed photoprobes, m-PAL-Cit-P and p-PAL-Cit-P, for identifying previously unidentified Dol-P-interacting proteins is supported by the observation that they are enzymatically mannosylated by Man-P-Dol synthase (MPDS) from Chinese Hamster Ovary (CHO) cells at an enzymatic rate similar to that for Dol-P. MS analyses reveal that DPM1, ALG14 and several other yeast ER proteins involved in DLO biosynthesis and lipid-mediated protein O-mannosylation photoreacted with the novel probes. CONCLUSION: The newly-designed photoprobes described in this paper provide promising new tools for the identification of yet to be identified Dol-P interacting ER proteins in yeast and mammalian cells, including the Dol-P flippase required for the "re-cycling" of the glycosyl carrier lipid from the lumenal monolayer of the ER to the cytoplasmic leaflet for new rounds of DLO synthesis.

9.
Bioorg Med Chem Lett ; 24(18): 4414-4417, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25150376

RESUMEN

An efficient, diversity oriented synthesis of homoisoprenoid α-monofluorophosphonates utilizing electrophilic fluorination is presented along with their activity as inhibitors of PPAPDC2 family integral membrane lipid phosphatases. These novel phosphatase-resistant analogues of isoprenoid monophosphates are a platform for further structure-activity relationship studies and provide access to other isoprenoid family members where the phosphate ester oxygen is replaced by a α-monofluoromethylene moiety.


Asunto(s)
Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Fosfatos de Poliisoprenilo/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Estructura Molecular , Monoéster Fosfórico Hidrolasas/metabolismo , Fosfatos de Poliisoprenilo/síntesis química , Fosfatos de Poliisoprenilo/química , Relación Estructura-Actividad
10.
PLoS One ; 9(2): e89892, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24587105

RESUMEN

Dysregulation of Ras and Rho family small GTPases drives the invasion and metastasis of multiple cancers. For their biological functions, these GTPases require proper subcellular localization to cellular membranes, which is regulated by a series of post-translational modifications that result in either farnesylation or geranylgeranylation of the C-terminal CAAX motif. This concept provided the rationale for targeting farnesyltransferase (FTase) and geranylgeranyltransferases (GGTase) for cancer treatment. However, the resulting prenyl transferase inhibitors have not performed well in the clinic due to issues with alternative prenylation and toxicity. As an alternative, we have developed a unique class of potential anti-cancer therapeutics called Prenyl Function Inhibitors (PFIs), which are farnesol or geranyl-geraniol analogs that act as alternate substrates for FTase or GGTase. Here, we test the ability of our lead PFIs, anilinogeraniol (AGOH) and anilinofarnesol (AFOH), to block the invasion of breast cancer cells. We found that AGOH treatment effectively decreased invasion of MDA-MB-231 cells in a two-dimensional (2D) invasion assay at 100 µM while it blocked invasive growth in three-dimensional (3D) culture model at as little as 20 µM. Notably, the effect of AGOH on 3D invasive growth was phenocopied by electroporation of cells with C3 exotransferase. To determine if RhoA and RhoC were direct targets of AGOH, we performed Rho activity assays in MDA-MB-231 and MDA-MB-468 cells and found that AGOH blocked RhoA and RhoC activation in response to LPA and EGF stimulation. Notably, the geranylgeraniol analog AFOH was more potent than AGOH in inhibiting RhoA and RhoC activation and invasive growth. Interestingly, neither AGOH nor AFOH impacted 3D growth of MCF10A cells. Collectively, this study demonstrates that AGOH and AFOH dramatically inhibit breast cancer invasion, at least in part by blocking Rho function, thus, suggesting that targeting prenylation by using PFIs may offer a promising mechanism for treatment of invasive breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Invasividad Neoplásica/prevención & control , Prenilación de Proteína/efectos de los fármacos , Terpenos/farmacología , Proteínas de Unión al GTP rho/metabolismo , Monoterpenos Acíclicos , Antineoplásicos/metabolismo , Técnicas de Cultivo de Célula , Farnesol/farmacología , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Estructura Molecular , Terpenos/metabolismo
11.
Mol Cell Biochem ; 387(1-2): 177-86, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24194124

RESUMEN

Mutations in Ras isoforms such as K-Ras, N-Ras, and H-Ras contribute to roughly 85, 15, and 1% of human cancers, respectively. Proper membrane targeting of these Ras isoforms, a prerequisite for Ras activity, requires farnesylation or geranylgeranylation at the C-terminal CAAX box. We devised an in vivo screening strategy based on monitoring Ras activation and phenotypic physiological outputs for assaying synthetic Ras function inhibitors (RFI). Ras activity was visualized by the translocation of RBD Raf1 -GFP to activated Ras at the plasma membrane. By using this strategy, we screened one synthetic farnesyl substrate analog (AGOH) along with nine putative inhibitors and found that only m-CN-AGOH inhibited Ras activation. Phenotypic analysis of starving cells could be used to monitor polarization, motility, and the inability of these treated cells to aggregate properly during fruiting body formation. Incorporation of AGOH and m-CN-AGOH to cellular proteins was detected by western blot. These screening assays can be incorporated into a high throughput screening format using Dictyostelium discoideum and automated microscopy to determine effective RFIs. These RFI candidates can then be further tested in mammalian systems.


Asunto(s)
Compuestos de Anilina/farmacología , Antineoplásicos/farmacología , Monoterpenos/farmacología , Proteínas ras/metabolismo , Polaridad Celular/efectos de los fármacos , Dictyostelium/citología , Dictyostelium/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Activación Enzimática , Ensayos Analíticos de Alto Rendimiento , Humanos , Fenotipo , Proteínas ras/antagonistas & inhibidores
12.
J Labelled Comp Radiopharm ; 56(8): 370-5, 2013 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-24285475

RESUMEN

A Wittig reaction employing Li(CD3)2CP(C6H5)3 was used to prepare d6-farnesol and d6-geranylgeraniol. Reductive amination of aniline-2,3,4,5,6-d5 was used to prepare the unnatural isoprenoid analogues d5-anilinogeraniol and d5-anilinofarnesol. All of these deuterated isoprenols were elaborated into their diphosphate and cysteine thioether derivatives suitable for use as stable-isotope labeled standards for quantitative mass spectrometric analysis.


Asunto(s)
Cisteína/análogos & derivados , Deuterio/síntesis química , Fosfatos de Poliisoprenilo/síntesis química , Cisteína/síntesis química , Marcaje Isotópico , Espectrometría de Masas/métodos
13.
J Biol Chem ; 288(38): 27444-27455, 2013 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-23908355

RESUMEN

Mammalian cells can use exogenous isoprenols to generate isoprenoid diphosphate substrates for protein isoprenylation, but the mechanism, efficiency, and biological importance of this process are not known. We developed mass spectrometry-based methods using chemical probes and newly synthesized stable isotope-labeled tracers to quantitate incorporation of exogenously provided farnesol, geranylgeraniol, and unnatural analogs of these isoprenols containing an aniline group into isoprenoid diphosphates and protein isoprenylcysteines by cultured human cancer cell lines. We found that at exogenous isoprenol concentrations >10 µM, this process can generate as much as 50% of the cellular isoprenoid diphosphate pool used for protein isoprenylation. Mutational activation of p53 in MDA-MB-231 breast cancer cells up-regulates the mevalonate pathway to promote tumor invasiveness. p53 silencing or pharmacological inhibition of HMG-CoA reductase in these cells decreases protein isoprenylation from endogenously synthesized isoprenoids but enhances the use of exogenous isoprenols for this purpose, indicating that this latter process is regulated independently of the mevalonate pathway. Our observations suggest unique opportunities for design of cancer cell-directed therapies and may provide insights into mechanisms underlying pleiotropic therapeutic benefits and unwanted side effects of mevalonate pathway inhibition.


Asunto(s)
Antineoplásicos/farmacología , Diterpenos/farmacología , Farnesol/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Ácido Mevalónico/metabolismo , Neoplasias/tratamiento farmacológico , Fosfatos de Poliisoprenilo/metabolismo , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Diterpenos/farmacocinética , Farnesol/análogos & derivados , Farnesol/farmacocinética , Humanos , Hidroximetilglutaril-CoA Reductasas/genética , Hidroximetilglutaril-CoA Reductasas/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacocinética , Mutación , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Prenilación de Proteína/efectos de los fármacos , Prenilación de Proteína/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
14.
Synlett ; 23(17): 2539-2543, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23125482

RESUMEN

A solid-phase three-component Huisgen reaction has been used to generate polar farnesol and farnesyl diphosphate analogues. The Cu(I)-catalyzed 1,3-cycloadditions of various azides with solid supported (E)-3-methylhept-2-en-6-yn-1-ol provided only the 1,4-disubstituted 1,2,3-triazole regioisomers. The organic azides were generated in situ to minimize handling of potentially explosive azides. We have employed this powerful 'click chemistry' to make farnesol analogues where both ß- and γ-isoprenes were replaced by triazole and substituted aromatic rings, respectively.

15.
Biochemistry ; 51(41): 8307-19, 2012 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-22989235

RESUMEN

Farnesylation is an important post-translational modification essential for the proper localization and function of many proteins. Transfer of the farnesyl group from farnesyl diphosphate (FPP) to proteins is catalyzed by protein farnesyltransferase (FTase). We employed a library of FPP analogues with a range of aryl groups substituting for individual isoprene moieties to examine some of the structural and electronic properties of the transfer of an analogue to the peptide catalyzed by FTase. Analysis of steady-state kinetics for modification of peptide substrates revealed that the multiple-turnover activity depends on the analogue structure. Analogues in which the first isoprene is replaced with a benzyl group and an analogue in which each isoprene is replaced with an aryl group are good substrates. In sharp contrast with the steady-state reaction, the single-turnover rate constant for dansyl-GCVLS alkylation was found to be the same for all analogues, despite the increased chemical reactivity of the benzyl analogues and the increased steric bulk of other analogues. However, the single-turnover rate constant for alkylation does depend on the Ca(1)a(2)X peptide sequence. These results suggest that the isoprenoid transition-state conformation is preferred over the inactive E·FPP·Ca(1)a(2)X ternary complex conformation. Furthermore, these data suggest that the farnesyl binding site in the exit groove may be significantly more selective for the farnesyl diphosphate substrate than the active site binding pocket and therefore might be a useful site for the design of novel inhibitors.


Asunto(s)
Transferasas Alquil y Aril/metabolismo , Fosfatos de Poliisoprenilo/metabolismo , Sesquiterpenos/metabolismo , Alquilación , Catálisis , Cinética , Espectroscopía de Resonancia Magnética , Fosfatos de Poliisoprenilo/química , Sesquiterpenos/química , Especificidad por Sustrato
16.
J Lipid Res ; 53(6): 1176-82, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22448028

RESUMEN

Protein farnesyltransferase (FTase) inhibitors, generally called "FTIs," block the farnesylation of prelamin A, inhibiting the biogenesis of mature lamin A and leading to an accumulation of prelamin A within cells. A recent report found that a GGTI, an inhibitor of protein geranylgeranyltransferase-I (GGTase-I), caused an exaggerated accumulation of prelamin A in the presence of low amounts of an FTI. This finding was interpreted as indicating that prelamin A can be alternately prenylated by GGTase-I and that inhibiting both protein prenyltransferases leads to more prelamin A accumulation than blocking FTase alone. Here, we tested an alternative hypothesis-GGTIs are not specific for GGTase-I, and they lead to prelamin A accumulation by inhibiting ZMPSTE24 (a zinc metalloprotease that converts farnesyl-prelamin A to mature lamin A). In our studies, commonly used GGTIs caused prelamin A accumulation in human fibroblasts, but the prelamin A in GGTI-treated cells exhibited a more rapid electrophoretic mobility than prelamin A from FTI-treated cells. The latter finding suggested that the prelamin A in GGTI-treated cells might be farnesylated (which would be consistent with the notion that GGTIs inhibit ZMPSTE24). Indeed, metabolic labeling studies revealed that the prelamin A in GGTI-treated fibroblasts is farnesylated. Moreover, biochemical assays of ZMPSTE24 activity showed that ZMPSTE24 is potently inhibited by a GGTI. Our studies show that GGTIs inhibit ZMPSTE24, leading to an accumulation of farnesyl-prelamin A. Thus, caution is required when interpreting the effects of GGTIs on prelamin A processing.


Asunto(s)
Transferasas Alquil y Aril/antagonistas & inhibidores , Proteínas de la Membrana/antagonistas & inhibidores , Metaloendopeptidasas/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Peptidomiméticos/farmacología , Inhibidores de Proteasas/farmacología , Precursores de Proteínas/metabolismo , Animales , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Lamina Tipo A , Ratones
17.
J Mol Med (Berl) ; 90(2): 149-61, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21915711

RESUMEN

Deregulation of Ras/ERK signaling in myeloid leukemias makes this pathway an interesting target for drug development. Myeloid leukemia cell lines were screened for idarubicin-induced apoptosis, cell-cycle progression, cell-cycle-dependent MAP kinase kinase (MEK-1/2) activation, and Top2 expression. Cell-cycle-dependent activation of MEK/ERK signaling was blocked using farnesyltransferase inhibitor (FTI) BMS-214,662 and dual prenyltransferase inhibitor (DPI) L-778,123 to disrupt Ras signaling. Idarubicin caused a G2/M cell-cycle arrest characterized by elevated diphosphorylated MEK-1/2 and Top2α expression levels. The FTI/DPIs elicited distinct effects on Ras signaling, protein prenylation, cell cycling and apoptosis. Combining these FTI/DPIs with idarubicin synergistically inhibited proliferation of leukemia cell lines, but the L-778,123+idarubicin combination exhibited synergistic growth inhibition over a greater range of drug concentrations. Interestingly, combined FTI/DPI treatment synergistically inhibited cell proliferation, induced apoptosis and nearly completely blocked protein prenylation. Inhibition of K-Ras expression by RNA interference or blockade of its post-translational prenylation led to increased BMS-214,662-induced apoptosis. Our results suggest that nearly complete inhibition of protein prenylation using an FTI + DPI combination is the most effective method to induce apoptosis and to block anthracycline-induced activation of ERK signaling.


Asunto(s)
Antígenos de Neoplasias/metabolismo , ADN-Topoisomerasas de Tipo II/metabolismo , Proteínas de Unión al ADN/metabolismo , Sinergismo Farmacológico , Leucemia Mieloide/metabolismo , Sistema de Señalización de MAP Quinasas , Prenilación de Proteína/efectos de los fármacos , Antraciclinas , Apoptosis/efectos de los fármacos , Benzodiazepinas/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Dimetilaliltranstransferasa/antagonistas & inhibidores , Farnesiltransferasa/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Idarrubicina/farmacología , Imidazoles/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas de Unión a Poli-ADP-Ribosa
18.
Hum Mol Genet ; 20(3): 436-44, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21088111

RESUMEN

Hutchinson-Gilford progeria syndrome (HGPS) is caused by a mutant prelamin A, progerin, that terminates with a farnesylcysteine. HGPS knock-in mice (Lmna(HG/+)) develop severe progeria-like disease phenotypes. These phenotypes can be ameliorated with a protein farnesyltransferase inhibitor (FTI), suggesting that progerin's farnesyl lipid is important for disease pathogenesis and raising the possibility that FTIs could be useful for treating humans with HGPS. Subsequent studies showed that mice expressing non-farnesylated progerin (Lmna(nHG/+) mice, in which progerin's carboxyl-terminal -CSIM motif was changed to -SSIM) also develop severe progeria, raising doubts about whether any treatment targeting protein prenylation would be particularly effective. We suspected that those doubts might be premature and hypothesized that the persistent disease in Lmna(nHG/+) mice could be an unanticipated consequence of the cysteine-to-serine substitution that was used to eliminate farnesylation. To test this hypothesis, we generated a second knock-in allele yielding non-farnesylated progerin (Lmna(csmHG)) in which the carboxyl-terminal -CSIM motif was changed to -CSM. We then compared disease phenotypes in mice harboring the Lmna(nHG) or Lmna(csmHG) allele. As expected, Lmna(nHG/+) and Lmna(nHG/nHG) mice developed severe progeria-like disease phenotypes, including osteolytic lesions and rib fractures, osteoporosis, slow growth and reduced survival. In contrast, Lmna(csmHG/+) and Lmna(csmHG/csmHG) mice exhibited no bone disease and displayed entirely normal body weights and survival. The frequencies of misshapen cell nuclei were lower in Lmna(csmHG/+) and Lmna(csmHG/csmHG) fibroblasts. These studies show that the ability of non-farnesylated progerin to elicit disease depends on the carboxyl-terminal mutation used to eliminate protein prenylation.


Asunto(s)
Farnesiltransferasa/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Progeria/genética , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Sustitución de Aminoácidos , Animales , Inhibidores Enzimáticos/farmacología , Farnesiltransferasa/antagonistas & inhibidores , Femenino , Técnicas de Sustitución del Gen , Imidazoles/farmacología , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Masculino , Ratones , Mutación , Proteínas Nucleares/química , Fenotipo , Progeria/metabolismo , Progeria/patología , Progeria/fisiopatología , Precursores de Proteínas/química , Prenilación de Proteína
19.
Anticancer Res ; 30(7): 2505-12, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20682975

RESUMEN

BACKGROUND: The KRAS oncogene has a high prevalence in solid malignancies. Targeting KRAS and inappropriate activation of the MAPK pathway with novel drugs is of interest. This study developed and screened a library of compounds designed to inhibit KRAS signaling by altering prenyl function. MATERIALS AND METHODS: To screen a library of novel farnesyl analogs for their anticancer activity in human lung cancer and breast cancer cell lines. To evaluate if the designed and actual pharmacology are congruent. RESULTS: Sixty-seven novel compounds were tested and 70% of them screened positive for activity in at least one cell line. Two active compounds inhibited phosphorylation of MAP kinase consistent with KRAS inhibition. CONCLUSION: Although 47 of the 67 novel agents screened positive for activity, none of them were highly potent. However, targeting RAS with compounds that compete with farnesyl and geranylgeranyl modification of the protein remains viable and further work is already underway to create second generation molecules.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Fosfatos de Poliisoprenilo/farmacología , Sesquiterpenos/farmacología , Terpenos/farmacología , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Transferasas Alquil y Aril/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Genes ras/efectos de los fármacos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas p21(ras) , Proteínas ras/metabolismo
20.
Hum Mol Genet ; 19(13): 2682-94, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20421363

RESUMEN

Lamin A is formed from prelamin A by four post-translational processing steps-farnesylation, release of the last three amino acids of the protein, methylation of the farnesylcysteine and the endoproteolytic release of the C-terminal 15 amino acids (including the farnesylcysteine methyl ester). When the final processing step does not occur, a farnesylated and methylated prelamin A accumulates in cells, causing a severe progeroid disease, restrictive dermopathy (RD). Whether RD is caused by the retention of farnesyl lipid on prelamin A, or by the retention of the last 15 amino acids of the protein, is unknown. To address this issue, we created knock-in mice harboring a mutant Lmna allele (LmnanPLAO) that yields exclusively non-farnesylated prelamin A (and no lamin C). These mice had no evidence of progeria but succumbed to cardiomyopathy. We suspected that the non-farnesylated prelamin A in the tissues of these mice would be strikingly mislocalized to the nucleoplasm, but this was not the case; most was at the nuclear rim (indistinguishable from the lamin A in wild-type mice). The cardiomyopathy could not be ascribed to an absence of lamin C because mice expressing an otherwise identical knock-in allele yielding only wild-type prelamin A appeared normal. We conclude that lamin C synthesis is dispensable in mice and that the failure to convert prelamin A to mature lamin A causes cardiomyopathy (at least in the absence of lamin C). The latter finding is potentially relevant to the long-term use of protein farnesyltransferase inhibitors, which lead to an accumulation of non-farnesylated prelamin A.


Asunto(s)
Cardiomiopatías/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Progeria/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Animales , Cardiomiopatías/etiología , Fibroblastos/metabolismo , Fibroblastos/patología , Técnicas de Sustitución del Gen , Lamina Tipo A/metabolismo , Ratones , Ratones Endogámicos , Modelos Animales , Progeria/etiología , Prenilación de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...