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











Intervalo de año de publicación
1.
Molecules ; 26(20)2021 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-34684845

RESUMEN

4-Hydroxycoumarin (4HC) has been used as a lead compound for the chemical synthesis of various bioactive substances and drugs. Its prenylated derivatives exhibit potent antibacterial, antitubercular, anticoagulant, and anti-cancer activities. In doing this, E. coli BL21(DE3)pLysS strain was engineered as the in vivo prenylation system to produce the farnesyl derivatives of 4HC by coexpressing the genes encoding Aspergillus terreus aromatic prenyltransferase (AtaPT) and truncated 1-deoxy-D-xylose 5-phosphate synthase of Croton stellatopilosus (CstDXS), where 4HC was the fed precursor. Based on the high-resolution LC-ESI(±)-QTOF-MS/MS with the use of in silico tools (e.g., MetFrag, SIRIUS (version 4.8.2), CSI:FingerID, and CANOPUS), the first major prenylated product (named compound-1) was detected and ultimately elucidated as ferulenol, in which information concerning the correct molecular formula, chemical structure, substructures, and classifications were obtained. The prenylated product (named compound-2) was also detected as the minor product, where this structure proposed to be the isomeric structure of ferulenol formed via the tautomerization. Note that both products were secreted into the culture medium of the recombinant E. coli and could be produced without the external supply of prenyl precursors. The results suggested the potential use of this engineered pathway for synthesizing the farnesylated-4HC derivatives, especially ferulenol.


Asunto(s)
Cumarinas/metabolismo , Escherichia coli/metabolismo , 4-Hidroxicumarinas/metabolismo , Aspergillus/metabolismo , Simulación por Computador , Dimetilaliltranstransferasa/metabolismo , Cinética , Prenilación/fisiología
2.
PLoS Pathog ; 17(10): e1009726, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34695163

RESUMEN

The zinc finger antiviral protein (ZAP) is a broad inhibitor of virus replication. Its best-characterized function is to bind CpG dinucleotides present in viral RNAs and, through the recruitment of TRIM25, KHNYN and other cofactors, target them for degradation or prevent their translation. The long and short isoforms of ZAP (ZAP-L and ZAP-S) have different intracellular localization and it is unclear how this regulates their antiviral activity against viruses with different sites of replication. Using ZAP-sensitive and ZAP-insensitive human immunodeficiency virus type I (HIV-1), which transcribe the viral RNA in the nucleus and assemble virions at the plasma membrane, we show that the catalytically inactive poly-ADP-ribose polymerase (PARP) domain in ZAP-L is essential for CpG-specific viral restriction. Mutation of a crucial cysteine in the C-terminal CaaX box that mediates S-farnesylation and, to a lesser extent, the residues in place of the catalytic site triad within the PARP domain, disrupted the activity of ZAP-L. Addition of the CaaX box to ZAP-S partly restored antiviral activity, explaining why ZAP-S lacks antiviral activity for CpG-enriched HIV-1 despite conservation of the RNA-binding domain. Confocal microscopy confirmed the CaaX motif mediated localization of ZAP-L to vesicular structures and enhanced physical association with intracellular membranes. Importantly, the PARP domain and CaaX box together jointly modulate the interaction between ZAP-L and its cofactors TRIM25 and KHNYN, implying that its proper subcellular localisation is required to establish an antiviral complex. The essential contribution of the PARP domain and CaaX box to ZAP-L antiviral activity was further confirmed by inhibition of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication, which replicates in double-membrane vesicles derived from the endoplasmic reticulum. Thus, compartmentalization of ZAP-L on intracellular membranes provides an essential effector function in ZAP-L-mediated antiviral activity against divergent viruses with different subcellular replication sites.


Asunto(s)
Prenilación/fisiología , Virus ARN/efectos de los fármacos , Proteínas de Unión al ARN/farmacología , Replicación Viral/fisiología , Islas de CpG/fisiología , Células HEK293 , VIH-1/fisiología , Células HeLa , Humanos , Virus ARN/fisiología , ARN Viral/química , ARN Viral/metabolismo , Motivos de Unión al ARN/fisiología , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , SARS-CoV-2/fisiología , Transfección , Replicación Viral/efectos de los fármacos
3.
Int J Mol Sci ; 22(11)2021 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-34067346

RESUMEN

Prenylated flavonoids are an important class of naturally occurring flavonoids with important biological activity, but their low abundance in nature limits their application in medicines. Here, we showed the hemisynthesis and the determination of various biological activities of seven prenylated flavonoids, named 7-13, with an emphasis on antimicrobial ones. Compounds 9, 11, and 12 showed inhibitory activity against human pathogenic fungi. Compounds 11, 12 (flavanones) and 13 (isoflavone) were the most active against clinical isolated Staphylococcus aureus MRSA, showing that structural requirements as prenylation at position C-6 or C-8 and OH at positions C-5, 7, and 4' are key to the antibacterial activity. The combination of 11 or 12 with commercial antibiotics synergistically enhanced the antibacterial activity of vancomycin, ciprofloxacin, and methicillin in a factor of 10 to 100 times against drug-resistant bacteria. Compound 11 combined with ciprofloxacin was able to decrease the levels of ROS generated by ciprofloxacin. According to docking results of S enantiomer of 11 with ATP-binding cassette transporter showed the most favorable binding energy; however, more studies are needed to support this result.


Asunto(s)
Antibacterianos/farmacología , Flavonoides/farmacología , Prenilación/fisiología , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Simulación por Computador , Flavanonas/farmacología , Hongos/efectos de los fármacos , Humanos , Isoflavonas/farmacología , Ratones , Pruebas de Sensibilidad Microbiana/métodos , Especies Reactivas de Oxígeno/metabolismo , Staphylococcus aureus/efectos de los fármacos
4.
Int J Biol Macromol ; 174: 61-68, 2021 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-33493569

RESUMEN

This study was to assess the possibility of using competitive and slow binding experiments with affinity-based ultrafiltration UPLC-QTof-MS analysis to identify potent bacterial neuraminidase (bNA) inhibitors from the Broussonetia papyrifera roots extract. To isolate unbound compounds from the enzyme-binding complex, the root bark extracts were either incubated in the absence of bNA, in the presence of bNA, or with the time-dependent bNA before the ultrafiltration was performed. Thirteen flavonoids were separated from the target extract, and their inhibitory activities were tested against bNA. The isolated flavonoids exhibited potent inhibition against NA (IC50 = 0.7-54.0 µM). Our kinetic analysis of representative active flavonoids (1, 2, and 6) showed slow and time-dependent reversible inhibition. Additionally, chalcones exhibited noncompetitive inhibition characteristics, whereas flavonols and flavans showed mixed-type behavior. The computational results supported the experimental behaviors of flavonoids 2, 6, 10, and 12, indicating that bounded to the active site, but flavonoids 6 and 10 binds near but not accurately at the active site. Although this is mixed-type inhibition, their binding can be considered competitive.


Asunto(s)
Broussonetia/química , Flavonoides/química , Raíces de Plantas/química , Chalcona/química , Chalconas/química , Flavonoles/química , Cinética , Neuraminidasa/química , Neuraminidasa/aislamiento & purificación , Neuraminidasa/metabolismo , Corteza de la Planta/química , Extractos Vegetales/química , Polifenoles/química , Prenilación/fisiología
5.
PLoS One ; 15(12): e0239269, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33315887

RESUMEN

The integral membrane zinc metalloprotease ZMPSTE24 plays a key role in the proteolytic processing of farnesylated prelamin A, the precursor of the nuclear scaffold protein lamin A. Failure of this processing step results in the accumulation of permanently farnesylated forms of prelamin A which cause the premature aging disease Hutchinson-Gilford Progeria Syndrome (HGPS), as well as related progeroid disorders, and may also play a role in physiological aging. ZMPSTE24 is an intriguing and unusual protease because its active site is located inside of a closed intramembrane chamber formed by seven transmembrane spans with side portals in the chamber permitting substrate entry. The specific features of prelamin A that make it the sole known substrate for ZMPSTE24 in mammalian cells are not well-defined. At the outset of this work it was known that farnesylation is essential for prelamin A cleavage in vivo and that the C-terminal region of prelamin A (41 amino acids) is sufficient for recognition and processing. Here we investigated additional features of prelamin A that are required for cleavage by ZMPSTE24 using a well-established humanized yeast system. We analyzed the 14-residue C-terminal region of prelamin A that lies between the ZMPSTE24 cleavage site and the farnesylated cysteine, as well 23-residue region N-terminal to the cleavage site, by generating a series of alanine substitutions, alanine additions, and deletions in prelamin A. Surprisingly, we found that there is considerable flexibility in specific requirements for the length and composition of these regions. We discuss how this flexibility can be reconciled with ZMPSTE24's selectivity for prelamin A.


Asunto(s)
Lamina Tipo A/metabolismo , Membranas/metabolismo , Metaloendopeptidasas/metabolismo , Metaloproteasas/metabolismo , Zinc/metabolismo , Alanina/metabolismo , Secuencia de Aminoácidos , Núcleo Celular/metabolismo , Cisteína/metabolismo , Proteínas de la Membrana/metabolismo , Prenilación/fisiología , Levaduras/metabolismo
6.
J Ethnopharmacol ; 263: 113147, 2020 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-32736058

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Macaranga Thou. (Euphorbiaceae) is a large genus that comprises over 300 species distributed between Western Africa and the islands of the South Pacific. Plants of this genus have a long-standing history of use in traditional medicine for different purposes, including the treatment of inflammation. Fresh and dried leaves of certain Macaranga species (e.g. M. tanarius (L.) Müll.Arg.), have been used to treat cuts, bruises, boils, swellings, sores and covering of wounds in general. Several reports described Macaranga spp. being a rich source of polyphenols, such as prenylated stilbenoids and flavonoids, mostly responsible for its biological activity. Similarly, an abundant content of prenylated stilbenes was also described in M. siamensis S.J.Davies, species recently identified (2001) in Thailand. While the respective biological activity of the prenylated stilbenes from M. siamensis was poorly investigated to date, our recent study pointed out the interest as the natural source of several novel anti-inflammatory stilbenoids isolated from this species. AIM OF THE STUDY: This work investigated the potential anti-inflammatory effects of the stilbenoid macasiamenene F (MF) isolated from M. siamensis S.J.Davies (Euphorbiaceae) on the lipopolysaccharide (LPS)-induced inflammation-like response of monocytes and microglia, major cells involved in the peripheral and central inflammatory response, respectively. MATERIALS AND METHODS: LPS-induced stimulation of TLR4 signaling led to the activation of inflammatory pathways in in vitro models of THP-1 and THP-1-XBlue™-MD2-CD14 human monocytes, BV-2 mouse microglia, and an ex vivo model of brain-sorted mouse microglia. The ability of the stilbenoid MF to intervene in the IкB/NF-кB and MAPKs/AP-1 inflammatory cascade was investigated. The gene and protein expressions of the pro-inflammatory cytokines IL-1ß and TNF-α were evaluated at the transcription and translation levels. The protective effect of MF against LPS-triggered microglial loss was assessed by cell counting and the LDH assay. RESULTS: MF demonstrated beneficial effects, reducing both monocyte and microglial inflammation as assessed in vitro. It efficiently inhibited the degradation of IкBα, thereby reducing the NF-кB activity and TNF-α expression in human monocytes. Furthermore, the LPS-induced expression of IL-1ß and TNF-α in microglia was dampened by pre-, co-, or post-treatment with MF. In addition to its anti-inflammatory effect, MF demonstrated a cytoprotective effect against the LPS-induced death of BV-2 microglia. CONCLUSION: Our research into anti-inflammatory and protective effects of MF has shown that it is a promising candidate for further in vitro and in vivo investigations of MF interventions with respect to acute and chronic inflammation, including potentially beneficial effects on the inflammatory component of brain diseases such as stroke and Alzheimer's disease.


Asunto(s)
Antiinflamatorios/uso terapéutico , Citoprotección/efectos de los fármacos , Euphorbiaceae , Microglía/efectos de los fármacos , Monocitos/efectos de los fármacos , Prenilación/efectos de los fármacos , Estilbenos/uso terapéutico , Animales , Antiinflamatorios/aislamiento & purificación , Antiinflamatorios/farmacología , Línea Celular Tumoral , Células Cultivadas , Citoprotección/fisiología , Relación Dosis-Respuesta a Droga , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Monocitos/metabolismo , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Prenilación/fisiología , Estilbenos/aislamiento & purificación , Estilbenos/farmacología
7.
Biochem Pharmacol ; 162: 109-122, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30316820

RESUMEN

The development of neuroprotective agents is necessary for the treatment of neurodegenerative diseases. Here, we report PQA-11, a prenylated quinolinecarboxylic acid (PQA) derivative, as a potent neuroprotectant. PQA-11 inhibits glutamate-induced cell death and caspase-3 activation in hippocampal cultures, as well as inhibits N-Methyl-4-phenylpyridinium iodide- and amyloid ß1-42-induced cell death in SH-SY5Y cells. PQA-11 also suppresses mitogen-activated protein kinase kinase 4 (MKK4) and c-jun N-terminal kinase (JNK) signaling activated by these neurotoxins. Quartz crystal microbalance analysis and in vitro kinase assay reveal that PQA-11 interacts with MKK4, and inhibits its sphingosine-induced activation. The administration of PQA-11 by intraperitoneal injection alleviates 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced degeneration of nigrostriatal dopaminergic neurons in mice. These results suggest that PQA-11 is a unique MKK4 inhibitor with potent neuroprotective effects in vitro and in vivo. PQA-11 may be a valuable lead for the development of novel neuroprotectants.


Asunto(s)
Ácidos Carboxílicos/farmacología , MAP Quinasa Quinasa 4/antagonistas & inhibidores , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Prenilación/efectos de los fármacos , Quinolinas/farmacología , Animales , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Línea Celular Tumoral , Humanos , MAP Quinasa Quinasa 4/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Prenilación/fisiología
8.
Proc Natl Acad Sci U S A ; 115(38): 9563-9568, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30190425

RESUMEN

SmgGDS has dual functions in cells and regulates small GTPases as both a guanine nucleotide exchange factor (GEF) for the Rho family and a molecular chaperone for small GTPases possessing a C-terminal polybasic region followed by four C-terminal residues called the CaaX motif, which is posttranslationally prenylated at its cysteine residue. Our recent structural work revealed that SmgGDS folds into tandem copies of armadillo-repeat motifs (ARMs) that are not present in other GEFs. However, the precise mechanism of GEF activity and recognition mechanism for the prenylated CaaX motif remain unknown because SmgGDS does not have a typical GEF catalytic domain and lacks a pocket to accommodate a prenyl group. Here, we aimed to determine the crystal structure of the SmgGDS/farnesylated RhoA complex. We found that SmgGDS induces a significant conformational change in the switch I and II regions that opens up the nucleotide-binding site, with the prenyl group fitting into the cryptic pocket in the N-terminal ARMs. Taken together, our findings could advance the understanding of the role of SmgGDS and enable drug design strategies for targeting SmgGDS and small GTPases.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/química , Chaperonas Moleculares/química , Proteínas de Unión al GTP Monoméricas/metabolismo , Pliegue de Proteína , Proteína de Unión al GTP rhoA/química , Secuencias de Aminoácidos , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Diseño de Fármacos , Pruebas de Enzimas , Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Chaperonas Moleculares/metabolismo , Simulación del Acoplamiento Molecular , Prenilación/fisiología , Unión Proteica , Proteína de Unión al GTP rhoA/metabolismo
9.
Plant Signal Behav ; 12(10): e1382795, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28949830

RESUMEN

Protein farnesylation refers to the addition of a 15-carbon farnesyl isoprenoid to the cysteine residue of the CaaX motif at the carboxy terminus of target proteins. In spite of its known roles in plant development and abiotic stress tolerance, how these processes are precisely regulated by farnesylation had remained elusive. We recently showed that CYP85A2, the cytochrome P450, which converts castasterone to brassinolide in the last step of brassinosteroid synthesis must be farnesylated in order to function in this pathway. Lack of either CYP85A2 or the farnesylation motif of CYP85A2 resulted in reduced brassinolide accumulation, hypersensitivity to ABA, and increased plant drought tolerance. In this study, we have assessed the influence of the N-terminal secretory signal and the C-terminal CaaX motif of CYP85A2 in mediating CYP85A2 function and targeting to endomembrane compartments. We show that CaaX motif could still target CYPA85A2 in the absence of an intact N-terminal secretory signal to the respective membrane compartments and partially rescue cyp85a2-2 phenotypes. However, in the absence of both the CaaX motif and the secretory signal, CYP85A2 is not targeted to the membranes and becomes unstable.


Asunto(s)
Ácido Abscísico/metabolismo , Proteínas de Arabidopsis/metabolismo , Brasinoesteroides/metabolismo , Semillas/metabolismo , Esteroides Heterocíclicos/metabolismo , Proteínas de Arabidopsis/genética , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Prenilación/genética , Prenilación/fisiología , Semillas/genética
10.
Arch Pharm Res ; 40(1): 32-36, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25749845

RESUMEN

Versicolols A and B (1 and 2), two rare prenylated isocoumarin derivatives, along with five known isocoumarins (3-7) were isolated from the fermentation products of an endophytic fungus Aspergillus versicolor. Their structures were elucidated on the basis of extensive spectroscopic analysis, including 1D- and 2D-NMR techniques. Compounds 1 and 2 were evaluated for their cytotoxicity against five human tumor cell lines. The results showed that compounds 1 exhibited weak cytotoxicity against A549 and MCF7 cells with IC50 values of 9.4 and 8.8 µm, and compound 2 exhibited weak cytotoxicity against SHSY5Y and MCF7 cells with IC50 values of 8.2 and 6.8 µm, respectively.


Asunto(s)
Aspergillus , Citotoxinas/química , Citotoxinas/metabolismo , Endófitos , Isocumarinas/química , Isocumarinas/metabolismo , Prenilación/fisiología , Células A549 , Citotoxinas/aislamiento & purificación , Humanos , Isocumarinas/aislamiento & purificación , Células MCF-7
11.
Biochem Biophys Res Commun ; 474(3): 594-598, 2016 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-27086854

RESUMEN

Increased activity of prenyl transferases is observed in pathological states of insulin resistance, diabetes, and obesity. Thus, functional inhibitors of farnesyl transferase (FTase) and geranylgeranyl transferase (GGTase) may be promising therapeutic treatments. We previously identified insulin responsive genes from a rat H4IIE hepatoma cell cDNA library, including ß-actin, EGR1, Pip92, c-fos, and Hsp60. In the present study, we investigated whether acute treatment with FTase and GGTase inhibitors would alter insulin responsive gene initiation and/or elongation rates. We observed differential regulation of insulin responsive gene expression, suggesting a differential sensitivity of these genes to one or both of the specific protein prenylation inhibitors.


Asunto(s)
Hepatocitos/metabolismo , Insulina/farmacología , Prenilación/fisiología , Proteínas/metabolismo , ARN Mensajero/metabolismo , Transcripción Genética/fisiología , Actinas/metabolismo , Animales , Línea Celular , Chaperonina 60/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Hepatocitos/efectos de los fármacos , Proteínas Mitocondriales/metabolismo , Prenilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Transcripción Genética/efectos de los fármacos
12.
Biochem Pharmacol ; 105: 55-65, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26827943

RESUMEN

Development of new immunosuppressing agents is necessary in organ transplantation or immune diseases. Because Ppc-1 exhibits a suppressing effect on interleukin-2 (IL2) production in Jurkat cells, we synthesized and screened Ppc-1 derivatives that preserve prenylated quinolinecarboxylic acid (PQA) structure, and identified compound 18 (PQA-18) as a novel molecule with immunosuppressing effect. PQA-18 suppressed not only IL2 but also IL4, IL6, and tumor necrosis factor-α production in human peripheral lymphocytes without affecting cell viability. Two-dimensional gel electrophoresis analysis and in vitro kinase assay revealed that PQA-18 inhibits kinase activity of p21-activated kinase 2 (PAK2). Administration of PQA-18 by intraperitoneal injection suppressed the population of a subset of regulatory T cells and the immunoglobulin (Ig) production against T cell-dependent antigens in mice. Treatment with the PQA-18 ointment on Nc/Nga mice, a model of human atopic dermatitis, improved skin lesions and serum IgE levels. These results suggest that PQA-18 is a unique PAK2 inhibitor with potent immunosuppressing effects in vitro and in vivo. PQA-18 may be a valuable lead for the development of novel immunosuppressants.


Asunto(s)
Ácidos Carboxílicos/farmacología , Inmunidad Celular/fisiología , Inmunosupresores/farmacología , Prenilación/fisiología , Quinolinas/farmacología , Quinasas p21 Activadas/metabolismo , Animales , Ácidos Carboxílicos/química , Dermatitis/tratamiento farmacológico , Dermatitis/inmunología , Dermatitis/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/inmunología , Células Jurkat , Masculino , Ratones , Ratones Endogámicos C57BL , Prenilación/efectos de los fármacos , Prenilación/inmunología , Quinolinas/química , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Ácidos Triyodobenzoicos/farmacología , Quinasas p21 Activadas/antagonistas & inhibidores , Quinasas p21 Activadas/inmunología
13.
Oncogene ; 34(24): 3095-106, 2015 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-25109332

RESUMEN

Protein geranylgeranylation (GGylation) is an important biochemical process for many cellular signaling molecules. Previous studies have shown that GGylation is essential for cell survival in many types of cancer. However, the molecular mechanism mediating the cell survival effect remains elusive. In this report, we show that the Hippo pathway mediates GGylation-dependent cell proliferation and migration in breast cancer cells. Blockade of GGylation enhanced phosphorylation of Mst1/2 and Lats1, and inhibited YAP and TAZ activity and the Hippo-YAP/TAZ pathway-dependent transcription. The effect of GGylation blockade on inhibition of breast cancer cell proliferation and migration is dependent on the Hippo-YAP/TAZ signaling, in which YAP appears to regulate cell proliferation and TAZ to regulate cell migration. Furthermore, GGylation-dependent cell proliferation is correlated with the activity of YAP/TAZ in breast cancer cells. Finally, Gγ and RhoA are the GGylated proteins that may transduce GGylation signals to the Hippo-YAP/TAZ pathway. Taken together, our studies have demonstrated that the Hippo-YAP/TAZ pathway is essential for GGylation-dependent cancer cell proliferation and migration.


Asunto(s)
Neoplasias de la Mama/patología , Movimiento Celular , Proliferación Celular , Prenilación/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Atorvastatina , Benzamidas/farmacología , Neoplasias de la Mama/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Células HEK293 , Ácidos Heptanoicos/farmacología , Vía de Señalización Hippo , Humanos , Células MCF-7 , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Pirroles/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Células Tumorales Cultivadas
14.
Infect Immun ; 82(1): 72-82, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24126522

RESUMEN

Upon entry of Legionella pneumophila into amoebas and macrophages, host-mediated farnesylation of the AnkB effector enables its anchoring to the Legionella-containing vacuole (LCV) membrane. On the LCV, AnkB triggers docking of K(48)-linked polyubiquitinated proteins that are degraded by the host proteasomes to elevate cellular levels of amino acids needed for intracellular proliferation. Interference with AnkB function triggers L. pneumophila to exhibit a starvation response and differentiate into the nonreplicative phase in response to the basal levels of cellular amino acids that are not sufficient to power intracellular proliferation of L. pneumophila. Therefore, we have determined whether the biological function of AnkB is temporally and spatially triggered upon bacterial attachment to the host cell to circumvent a counterproductive bacterial differentiation into the nonreplicative phase upon bacterial entry. Here, we show that upon attachment of L. pneumophila to human monocyte-derived macrophages (hMDMs), the host farnesylation and ubiquitination machineries are recruited by the Dot/Icm system to the plasma membrane exclusively beneath sites of bacterial attachment. Transcription and injection of ankB is triggered by attached extracellular bacteria followed by rapid farnesylation and anchoring of AnkB to the cytosolic side of the plasma membrane beneath bacterial attachment, where K(48)-linked polyubiquitinated proteins are assembled and degraded by the proteasomes, leading to a rapid rise in the cellular levels of amino acids. Our data represent a novel strategy by an intracellular pathogen that triggers rapid nutritional remodeling of the host cell upon attachment to the plasma membrane, and as a result, a gratuitous surplus of cellular amino acids is generated to support proliferation of the incoming pathogen.


Asunto(s)
Aminoácidos/biosíntesis , Ancirinas/fisiología , Adhesión Bacteriana/fisiología , Interacciones Huésped-Patógeno , Legionella pneumophila/fisiología , Macrófagos/microbiología , Proteínas de Unión Periplasmáticas/fisiología , Amoeba/microbiología , Sitios de Ligazón Microbiológica/fisiología , Membrana Celular/fisiología , Células Cultivadas , Humanos , Legionella pneumophila/patogenicidad , Prenilación/fisiología , Ubiquitinación/fisiología , Vacuolas/microbiología
15.
Haematologica ; 98(1): 31-40, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22801964

RESUMEN

Despite advances in immunosuppressive regimens, acute graft-versus-host disease remains a frequent complication of allogeneic hematopoietic cell transplantation. Pathogenic donor T cells are dependent on correct attachment of small GTPases to the cell membrane, mediated by farnesyl- or geranylgeranyl residues, which, therefore, constitute potential targets for graft-versus-host disease prophylaxis. A mouse model was used to study the impact of a farnesyl-transferase inhibitor and a geranylgeranyl-transferase inhibitor on acute graft-versus-host disease, anti-cytomegalovirus T-cell responses and graft-versus-leukemia activity. Treatment of mice undergoing allogeneic hematopoietic cell transplantation with farnesyl-transferase inhibitor and geranylgeranyl-transferase inhibitor reduced the histological severity of graft-versus-host disease and prolonged survival significantly. Mechanistically, farnesyl-transferase inhibitor and geranylgeranyl-transferase inhibitor treatment resulted in reduced alloantigen-driven expansion of CD4 T cells. In vivo treatment led to increased thymic cellularity and polyclonality of the T-cell receptor repertoire by reducing thymic graft-versus-host disease. These effects were absent when squalene production was blocked. The farnesyl-transferase inhibitor and geranylgeranyl-transferase inhibitor did not compromise CD8 function against leukemia cells or reconstitution of T cells that were subsequently responsible for anti-murine cytomegalovirus responses. In summary, we observed an immunomodulatory effect of inhibitors of farnesyl-transferase and geranylgeranyl-transferase on graft-versus-host disease, with enhanced functional immune reconstitution. In the light of the modest toxicity of farnesyl-transferase inhibitors such as tipifarnib in patients and the potent reduction of graft-versus-host disease in mice, farnesyl-transferase and geranylgeranyl-transferase inhibitors could help to reduce graft-versus-host disease significantly without having a negative impact on immune reconstitution.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/metabolismo , Prenilación/fisiología , Prenilación de Proteína/fisiología , Animales , Trasplante de Médula Ósea/efectos adversos , Linfocitos T CD4-Positivos/efectos de los fármacos , Enfermedad Injerto contra Huésped/prevención & control , Efecto Injerto vs Leucemia/efectos de los fármacos , Efecto Injerto vs Leucemia/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Prenilación/efectos de los fármacos , Prenilación de Proteína/efectos de los fármacos , Quinolonas/farmacología , Quinolonas/uso terapéutico , Resultado del Tratamiento
16.
Proc Natl Acad Sci U S A ; 109(27): 10843-8, 2012 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-22711838

RESUMEN

Members of the RAS small GTPase family regulate cellular responses to extracellular stimuli by mediating the flux through downstream signal transduction cascades. RAS activity is strongly dependent on its subcellular localization and its nucleotide-binding status, both of which are modulated by posttranslational modification. We have determined that RAS is posttranslationally acetylated on lysine 104. Molecular dynamics simulations suggested that this modification affects the conformational stability of the Switch II domain, which is critical for the ability of RAS to interact with guanine nucleotide exchange factors. Consistent with this model, an acetylation-mimetic mutation in K-RAS4B suppressed guanine nucleotide exchange factor-induced nucleotide exchange and inhibited in vitro transforming activity. These data suggest that lysine acetylation is a negative regulatory modification on RAS. Because mutations in RAS family members are extremely common in cancer, modulation of RAS acetylation may constitute a therapeutic approach.


Asunto(s)
Genes ras/fisiología , Procesamiento Proteico-Postraduccional/fisiología , Proteínas ras/química , Proteínas ras/metabolismo , Acetilación , Animales , Células COS , Chlorocebus aethiops , Factores de Intercambio de Guanina Nucleótido/metabolismo , Guanosina Trifosfato/química , Guanosina Trifosfato/metabolismo , Células HEK293 , Células HeLa , Humanos , Lisina/metabolismo , Mutagénesis Sitio-Dirigida , Prenilación/fisiología , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína/fisiología , Relación Estructura-Actividad , Proteínas ras/genética
17.
J Biol Chem ; 287(27): 23128-40, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22605333

RESUMEN

The maternally imprinted Ras-related tumor suppressor gene DiRas3 is lost or down-regulated in more than 60% of ovarian and breast cancers. The anti-tumorigenic effect of DiRas3 is achieved through several mechanisms, including inhibition of cell proliferation, motility, and invasion, as well as induction of apoptosis and autophagy. Re-expression of DiRas3 in cancer cells interferes with the signaling through Ras/MAPK and PI3K. Despite intensive research, the mode of interference of DiRas3 with the Ras/RAF/MEK/ERK signal transduction is still a matter of speculation. In this study, we show that DiRas3 associates with the H-Ras oncogene and that activation of H-Ras enforces this interaction. Furthermore, while associated with DiRas3, H-Ras is able to bind to its effector protein C-RAF. The resulting multimeric complex consisting of DiRas3, C-RAF, and active H-Ras is more stable than the two protein complexes H-Ras·C-RAF or H-Ras·DiRas3, respectively. The consequence of this complex formation is a DiRas3-mediated recruitment and anchorage of C-RAF to components of the membrane skeleton, suppression of C-RAF/B-RAF heterodimerization, and inhibition of C-RAF kinase activity.


Asunto(s)
Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Proto-Oncogénicas c-raf/química , Proteínas Proto-Oncogénicas c-raf/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Citoesqueleto/metabolismo , Dimerización , Genes Supresores de Tumor/fisiología , Humanos , Complejos Multiproteicos/metabolismo , Prenilación/fisiología , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas de Unión al GTP rho/genética
18.
Pharm Biol ; 49(3): 314-8, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21281247

RESUMEN

CONTEXT: A newly discovered geranyl prenylated chalcone, semisynthesized from naturally occurring nymphaeol C, has the ability to inhibit the growth of CNS1 (glioblastoma) and 13-06 (malignant glioma) cells. A second-order regression model was established to predict the normalized cell viability of CNS1 and 13-06 cells. OBJECTIVE: The goal of this study is to evaluate the influence of prenylated chalcone on the glioblastoma and malignant glioma cell lines. For the first time, response surface methodology (RSM) has been introduced to perform a cell line study. MATERIALS AND METHODS: A newly discovered prenylated chalcone was used. This compound is a member of the flavonoid family and possesses a common phenylbenzopyrone structure. Two independent factors, including prenylated chalcone concentration and uptake time, were carefully evaluated by a 2² factorial design. RSM was introduced as a new method for CNS1 and 13-06 cell line studies. RESULTS: For CNS1 cells, the least inhibition uptake time was 20.7 h, and the least inhibition dose was 12.4 µg/ml. For 13-06 cells, the best inhibition uptake time was 26.2 h, and the least inhibition dose was 12.0 µg/ml. DISCUSSION AND CONCLUSION: The RSM model successfully predicted the normalized cell viability of CNS1 and 13-06 cells through the use of prenylated chalcone. The results obtained in this study will be useful for further studies on the use of prenylated chalcone.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Chalcona/metabolismo , Glioblastoma/metabolismo , Glioma/metabolismo , Prenilación/fisiología , Animales , Línea Celular Tumoral , Supervivencia Celular/fisiología , Chalcona/aislamiento & purificación , Flavanonas/uso terapéutico , Glioblastoma/patología , Glioma/patología
19.
Prostate ; 70(9): 971-81, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20135644

RESUMEN

BACKGROUND: Autophagy is a cellular process of degradation of macromolecules and organelles and activated under nutritional stress. Statins are a class of inhibitors of 3-hydroxyl-3-methylglutaryl coenzyme A (HMG-CoA) reductase, a key enzyme in synthesis of cholesterol. Epidemiological studies have shown that statin use decreases the incidence of advanced prostate cancer. We explored the idea that treatment of atorvastatin, a commonly prescribed statin for treatment of hypercholesterolemia, induces autophagy in prostate cancer cells. METHODS: The atorvastatin-induced autophagic process in prostate cancer PC3 cells was determined by detection of cellular level of LC3-II, an autophagosomal marker, via immunoblotting and immunofluorescent staining. RESULTS: Atorvastatin treatment of PC3 cells for 40 hrs increased expression of LC3-II by more than 10 fold in a dose-dependent manner. Treatment of the cells with pepstatin A and E64-d, the autophagic protease inhibitors, dramatically increased atorvastatin-dependent LC3-II expression level, suggesting that atorvastatin induces autophagic flux. In addition, atorvastatin treatment caused rapid death of PC3 cells. Atorvastatin-induced autophagy and rapid cell death were reversed by addition of geranylgeraniol, not farnesol, into culture medium, indicating that atorvastatin-mediated inhibition of geranylgeranyl biosynthesis causes autophagy and cell death. Furthermore, atorvastatin did not induce autophagy or cell death in normal prostate RWPE1 cells, and induced only a minor autophagic response in AR-positive prostate cancer LNCaP cells. CONCLUSIONS: Our studies demonstrate that statins induce autophagy and autophagy-associated cell death in PC3 cells, likely through inhibition of geranylgeranylation, and suggest that autophagic response to statins may partially underlie the protective effects of statins on prostate cancer progression. Importantly, these findings highlight additional mechanisms by which statins might be used for prostate cancer therapy.


Asunto(s)
Autofagia/efectos de los fármacos , Ácidos Heptanoicos/farmacología , Prenilación/efectos de los fármacos , Próstata/metabolismo , Pirroles/farmacología , Anticolesterolemiantes/farmacología , Atorvastatina , Autofagia/fisiología , Western Blotting , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Masculino , Prenilación/fisiología , Próstata/efectos de los fármacos
20.
Biochemistry ; 47(32): 8424-33, 2008 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-18636747

RESUMEN

G protein gamma-subunits are isoprenylated and carboxyl methylated at the C-terminal cysteine residue, and the set of the posttranslational modifications is indispensable for the function of the photoreceptor G protein transducin (Talpha/Tbetagamma). To explore farnesyl-mediated molecular interactions, we investigated molecular targets of a Tbetagamma analogue that was engineered to have a photoreactive farnesyl analogue, (3-azidophenoxy)geranyl (POG), covalently bound to the C-terminal cysteine of Tgamma. POG-modified Tbetagamma was further subjected to modification by methylation at the C-terminal carboxyl group, which copies a complete set of the known posttranscriptional modifications of Tbetagamma. Photoaffinity labeling experiment with the photoreactive Tbetagamma analogue in its free form indicated that the POG moiety of Tgamma interacted with Tbeta. In the trimeric Talpha/Tbetagamma complex, the POG moiety was cross-linked with Talpha in addition to concurrent affinity labeling of Tbeta. When photoreactive Tbetagamma was reconstituted with Talpha and light-activated rhodopsin (Rh*) in rod outer segment (ROS) membranes, the POG moiety interacted with not only Talpha and Tbeta but also Rh* and membrane phospholipids. The cross-linked phospholipid species was analyzed by ELISA employing a variety of lipid-binding probes, which revealed phosphatidylethanolamine (PE) and phosphatidylserine (PS) as selective phospholipids for POG interaction in the ROS membranes. These results demonstrate that the modifying group of Tgamma plays an active role in protein-protein and protein-membrane interactions and suggest that the farnesyl-PE/PS interaction may support the efficient formation of the signaling ternary complex between transducin and Rh*.


Asunto(s)
Prenilación/fisiología , Dominios y Motivos de Interacción de Proteínas , Transducina/química , Transducina/metabolismo , Animales , Bovinos , Marcación de Gen , Humanos , Metilación , Mutagénesis Sitio-Dirigida , Dominios y Motivos de Interacción de Proteínas/genética , Transducción de Señal/fisiología , Transducina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA