Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 23
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
bioRxiv ; 2023 Oct 23.
Article de Anglais | MEDLINE | ID: mdl-37986908

RÉSUMÉ

ATP-binding cassette (ABC) transporters expressed at the blood-brain barrier (BBB) impede delivery of therapeutic agents to the brain, including agents to treat neurodegenerative diseases and primary and metastatic brain cancers. Two transporters, P-glycoprotein (P-gp, ABCB1) and ABCG2, are highly expressed at the BBB and are responsible for the efflux of numerous clinically useful chemotherapeutic agents, including irinotecan, paclitaxel, and doxorubicin. Based on a previous mouse model, we have generated transgenic zebrafish in which expression of NanoLuciferase (NanoLuc) is controlled by the promoter of glial fibrillary acidic protein, leading to expression in zebrafish glia. To identify agents that disrupt the BBB, including inhibitors of ABCB1 and ABCG2, we identified NanoLuc substrates that are also transported by P-gp, ABCG2, and their zebrafish homologs. These substrates will elevate the amount of bioluminescent light produced in the transgenic zebrafish with BBB disruption. We transfected HEK293 cells with NanoLuc and either human ABCB1, ABCG2, or their zebrafish homologs Abcb4 or Abcg2a, respectively, and expressed at the zebrafish BBB. We evaluated the luminescence of ten NanoLuc substrates, then screened the eight brightest to determine which are most efficiently effluxed by the ABC transporters. We identified one substrate efficiently pumped out by ABCB1, two by Abcb4, six by ABCG2, and four by Abcg2a. These data will aid in the development of a transgenic zebrafish model of the BBB to identify novel BBB disruptors and should prove useful in the development of other animal models that use NanoLuc as a reporter.

2.
ACS Cent Sci ; 9(4): 719-732, 2023 Apr 26.
Article de Anglais | MEDLINE | ID: mdl-37122464

RÉSUMÉ

Aberrant kinase activity contributes to the pathogenesis of brain cancers, neurodegeneration, and neuropsychiatric diseases, but identifying kinase inhibitors that function in the brain is challenging. Drug levels in blood do not predict efficacy in the brain because the blood-brain barrier prevents entry of most compounds. Rather, assessing kinase inhibition in the brain requires tissue dissection and biochemical analysis, a time-consuming and resource-intensive process. Here, we report kinase-modulated bioluminescent indicators (KiMBIs) for noninvasive longitudinal imaging of drug activity in the brain based on a recently optimized luciferase-luciferin system. We develop an ERK KiMBI to report inhibitors of the Ras-Raf-MEK-ERK pathway, for which no bioluminescent indicators previously existed. ERK KiMBI discriminates between brain-penetrant and nonpenetrant MEK inhibitors, reveals blood-tumor barrier leakiness in xenograft models, and reports MEK inhibitor pharmacodynamics in native brain tissues and intracranial xenografts. Finally, we use ERK KiMBI to screen ERK inhibitors for brain efficacy, identifying temuterkib as a promising brain-active ERK inhibitor, a result not predicted from chemical characteristics alone. Thus, KiMBIs enable the rapid identification and pharmacodynamic characterization of kinase inhibitors suitable for treating brain diseases.

3.
Nat Chem Biol ; 19(6): 731-739, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-36759751

RÉSUMÉ

Bioluminescence imaging (BLI) allows non-invasive visualization of cells and biochemical events in vivo and thus has become an indispensable technique in biomedical research. However, BLI in the central nervous system remains challenging because luciferases show relatively poor performance in the brain with existing substrates. Here, we report the discovery of a NanoLuc substrate with improved brain performance, cephalofurimazine (CFz). CFz paired with Antares luciferase produces greater than 20-fold more signal from the brain than the standard combination of D-luciferin with firefly luciferase. At standard doses, Antares-CFz matches AkaLuc-AkaLumine/TokeOni in brightness, while occasional higher dosing of CFz can be performed to obtain threefold more signal. CFz should allow the growing number of NanoLuc-based indicators to be applied to the brain with high sensitivity. Using CFz, we achieve video-rate non-invasive imaging of Antares in brains of freely moving mice and demonstrate non-invasive calcium imaging of sensory-evoked activity in genetically defined neurons.


Sujet(s)
Imagerie diagnostique , Mesures de luminescence , Souris , Animaux , Mesures de luminescence/méthodes , Encéphale/imagerie diagnostique , Luciférine de luciole , Luciférines
4.
Nat Commun ; 13(1): 2073, 2022 04 19.
Article de Anglais | MEDLINE | ID: mdl-35440107

RÉSUMÉ

Modulation of protein abundance using tag-Targeted Protein Degrader (tTPD) systems targeting FKBP12F36V (dTAGs) or HaloTag7 (HaloPROTACs) are powerful approaches for preclinical target validation. Interchanging tags and tag-targeting degraders is important to achieve efficient substrate degradation, yet limited degrader/tag pairs are available and side-by-side comparisons have not been performed. To expand the tTPD repertoire we developed catalytic NanoLuc-targeting PROTACs (NanoTACs) to hijack the CRL4CRBN complex and degrade NanoLuc tagged substrates, enabling rapid luminescence-based degradation screening. To benchmark NanoTACs against existing tTPD systems we use an interchangeable reporter system to comparatively test optimal degrader/tag pairs. Overall, we find the dTAG system exhibits superior degradation. To align tag-induced degradation with physiology we demonstrate that NanoTACs limit MLKL-driven necroptosis. In this work we extend the tTPD platform to include NanoTACs adding flexibility to tTPD studies, and benchmark each tTPD system to highlight the importance of comparing each system against each substrate.


Sujet(s)
Référenciation , Protéine 1A de liaison au tacrolimus , Luciferases , Protéolyse , Protéine 1A de liaison au tacrolimus/génétique
5.
Viruses ; 14(3)2022 03 05.
Article de Anglais | MEDLINE | ID: mdl-35336942

RÉSUMÉ

Animal models recapitulating COVID-19 are critical to enhance our understanding of SARS-CoV-2 pathogenesis. Intranasally inoculated transgenic mice expressing human angiotensin-converting enzyme 2 under the cytokeratin 18 promoter (K18-hACE2) represent a lethal model of SARS-CoV-2 infection. We evaluated the clinical and virological dynamics of SARS-CoV-2 using two intranasal doses (104 and 106 PFUs), with a detailed spatiotemporal pathologic analysis of the 106 dose cohort. Despite generally mild-to-moderate pneumonia, clinical decline resulting in euthanasia or death was commonly associated with hypothermia and viral neurodissemination independent of inoculation dose. Neuroinvasion was first observed at 4 days post-infection, initially restricted to the olfactory bulb suggesting axonal transport via the olfactory neuroepithelium as the earliest portal of entry. Absence of viremia suggests neuroinvasion occurs independently of transport across the blood-brain barrier. SARS-CoV-2 tropism was neither restricted to ACE2-expressing cells (e.g., AT1 pneumocytes), nor inclusive of some ACE2-positive cell lineages (e.g., bronchiolar epithelium and brain vasculature). Absence of detectable ACE2 protein expression in neurons but overexpression in neuroepithelium suggest this as the most likely portal of neuroinvasion, with subsequent ACE2 independent lethal neurodissemination. A paucity of epidemiological data and contradicting evidence for neuroinvasion and neurodissemination in humans call into question the translational relevance of this model.


Sujet(s)
COVID-19 , SARS-CoV-2 , Angiotensin-converting enzyme 2 , Animaux , Humains , Kératine-18 , Melphalan , Souris , Souris transgéniques , SARS-CoV-2/génétique , Tropisme viral , Gammaglobulines
6.
Adv Genet (Hoboken) ; 3(1): 2100055, 2022 Mar.
Article de Anglais | MEDLINE | ID: mdl-36619349

RÉSUMÉ

Cancer cells produce heterogeneous extracellular vesicles (EVs) as mediators of intercellular communication. This study focuses on a novel method to image EV subtypes and their biodistribution in vivo. A red-shifted bioluminescence resonance energy transfer (BRET) EV reporter is developed, called PalmReNL, which allows for highly sensitive EV tracking in vitro and in vivo. PalmReNL enables the authors to study the common surface molecules across EV subtypes that determine EV organotropism and their functional differences in cancer progression. Regardless of injection routes, whether retro-orbital or intraperitoneal, PalmReNL positive EVs, isolated from murine mammary carcinoma cells, localized to the lungs. The early appearance of metastatic foci in the lungs of mammary tumor-bearing mice following multiple intraperitoneal injections of the medium and large EV (m/lEV)-enriched fraction derived from mammary carcinoma cells is demonstrated. In addition, the results presented here show that tumor cell-derived m/lEVs act on distant tissues through upregulating LC3 expression within the lung.

7.
Adv Sci (Weinh) ; 8(13): 2100424, 2021 07.
Article de Anglais | MEDLINE | ID: mdl-34540558

RÉSUMÉ

Herein, a set of optogenetic tools (designated LiPOP) that enable photoswitchable necroptosis and pyroptosis in live cells with varying kinetics, is introduced. The LiPOP tools allow reconstruction of the key molecular steps involved in these two non-apoptotic cell death pathways by harnessing the power of light. Further, the use of LiPOPs coupled with upconversion nanoparticles or bioluminescence is demonstrated to achieve wireless optogenetic or chemo-optogenetic killing of cancer cells in multiple mouse tumor models. LiPOPs can trigger necroptotic and pyroptotic cell death in cultured prokaryotic or eukaryotic cells and in living animals, and set the stage for studying the role of non-apoptotic cell death pathways during microbial infection and anti-tumor immunity.


Sujet(s)
Mort cellulaire , Optogénétique , Animaux , Escherichia coli , Cellules HeLa/métabolisme , Cellules HeLa/transplantation , Humains , Protéines et peptides de signalisation intracellulaire/métabolisme , Leucémies , Lumière , Souris , Nanoparticules , Nécroptose , Optogénétique/méthodes , Protéines de liaison aux phosphates/métabolisme , Protein kinases/métabolisme , Pyroptose
8.
J Photochem Photobiol B ; 216: 112128, 2021 Mar.
Article de Anglais | MEDLINE | ID: mdl-33529963

RÉSUMÉ

NanoLuc luciferase recently gained popularity due to its small size and superior bioluminescence performance. For in vivo imaging applications, NanoLuc has been limited by its substrate furimazine, which has low solubility and bioavailability. Herein, we compared the performances of recently reported NanoLuc luciferase substrates for in vivo imaging in mice. Two substrates with improved aqueous solubility, hydrofurimazine and fluorofurimazine, were evaluated along with three stabilized O-acetylated furimazine analogues, the hikarazines. All 5 analogues, when tested in vitro, displayed greater signal intensity and reaction duration, in comparison to the standard NanoLuc substrate, furimazine. The two best-performing analogues from the in vitro study were selected for further in vivo testing. The NanoLuc/fluorofurimazine pair demonstrated the highest bioluminescence intensity, post intravenous administration. It was found to be around 9-fold brighter compared to the NanoLuc/furimazine and 11-fold more intense than the NanoLuc/hikarazine-003 pair, with an average of 3-fold higher light emission when the substrate was injected intraperitoneally, in a subcutaneous model. Excitingly, despite the fact that NanoLuc/fluorofurimazine emits mostly blue light, we prove that cells trapped in mice lungs vasculature could be visualised via the NanoLuc/fluorofurimazine pair and compare the results to the AkaLuc/AkaLumine system. Therefore, among the tested analogues, fluorofurimazine enables higher substrate loading and improved optical imaging sensitivity in small animals, upgrading the use of NanoLuc derived bioluminescent systems for deep tissue imaging.


Sujet(s)
Luciferases/composition chimique , Luminescents/composition chimique , Poumon/imagerie diagnostique , Vaisseaux rétiniens/imagerie diagnostique , Animaux , Furanes/composition chimique , Cellules HEK293 , Humains , Imidazoles/composition chimique , Infections à lentivirus , Lumière , Luciferases/métabolisme , Luminescents/métabolisme , Mâle , Souris de lignée BALB C , Souris nude , Imagerie optique , Pyrazines/composition chimique , Solubilité , Relation structure-activité
9.
bioRxiv ; 2021 Sep 23.
Article de Anglais | MEDLINE | ID: mdl-33469581

RÉSUMÉ

Animal models recapitulating distinctive features of severe COVID-19 are critical to enhance our understanding of SARS-CoV-2 pathogenesis. Transgenic mice expressing human angiotensin-converting enzyme 2 (hACE2) under the cytokeratin 18 promoter (K18-hACE2) represent a lethal model of SARS-CoV-2 infection. The precise mechanisms of lethality in this mouse model remain unclear. Here, we evaluated the spatiotemporal dynamics of SARS-CoV-2 infection for up to 14 days post-infection. Despite infection and moderate pneumonia, rapid clinical decline or death of mice was invariably associated with viral neuroinvasion and direct neuronal injury (including brain and spinal neurons). Neuroinvasion was observed as early as 4 dpi, with virus initially restricted to the olfactory bulb supporting axonal transport via the olfactory neuroepithelium as the earliest portal of entry. No evidence of viremia was detected suggesting neuroinvasion occurs independently of entry across the blood brain barrier. SARS-CoV-2 tropism was not restricted to ACE2-expressing cells (e.g., AT1 pneumocytes), and some ACE2-positive lineages were not associated with the presence of viral antigen (e.g., bronchiolar epithelium and brain capillaries). Detectable ACE2 expression was not observed in neurons, supporting overexpression of ACE2 in the nasal passages and neuroepithelium as more likely determinants of neuroinvasion in the K18-hACE2 model. Although our work incites caution in the utility of the K18-hACE2 model to study global aspects of SARS-CoV-2 pathogenesis, it underscores this model as a unique platform for exploring the mechanisms of SARS-CoV-2 neuropathogenesis that may have clinical relevance acknowledging the growing body of evidence that suggests COVID-19 may result in long-standing neurologic consequences.

10.
Int J Mol Sci ; 21(16)2020 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-32824188

RÉSUMÉ

Reporter genes are used to visualize intracellular biological phenomena, including viral infection. Here we demonstrate bioluminescent imaging of viral infection using the NanoBiT system in combination with intraperitoneal injection of a furimazine analogue, hydrofurimazine. This recently developed substrate has enhanced aqueous solubility allowing delivery of higher doses for in vivo imaging. The small high-affinity peptide tag (HiBiT), which is only 11 amino-acids in length, was engineered into a clinically used oncolytic adenovirus, and the complementary large protein (LgBiT) was constitutively expressed in tumor cells. Infection of the LgBiT expressing cells with the HiBiT oncolytic virus will reconstitute NanoLuc in the cytosol of the cell, providing strong bioluminescence upon treatment with substrate. This new bioluminescent system served as an early stage quantitative viral transduction reporter in vitro and also in vivo in mice, for longitudinal monitoring of oncolytic viral persistence in infected tumor cells. This platform provides novel opportunities for studying the biology of viruses in animal models.


Sujet(s)
Furanes/pharmacocinétique , Imidazoles/pharmacocinétique , Luminescents/pharmacocinétique , Protéines luminescentes/génétique , Imagerie optique/méthodes , Pyrazines/pharmacocinétique , Maladies virales/imagerie diagnostique , Adenoviridae/génétique , Animaux , Lignée cellulaire tumorale , Furanes/administration et posologie , Cellules HEK293 , Humains , Imidazoles/administration et posologie , Injections péritoneales , Luminescents/administration et posologie , Protéines luminescentes/métabolisme , Mâle , Souris , Souris de lignée BALB C , Souris nude , Oligopeptides/génétique , Oligopeptides/métabolisme , Virus oncolytiques/génétique , Pyrazines/administration et posologie , Protéines recombinantes/génétique
11.
Nat Methods ; 17(8): 852-860, 2020 08.
Article de Anglais | MEDLINE | ID: mdl-32661427

RÉSUMÉ

Sensitive detection of two biological events in vivo has long been a goal in bioluminescence imaging. Antares, a fusion of the luciferase NanoLuc to the orange fluorescent protein CyOFP, has emerged as a bright bioluminescent reporter with orthogonal substrate specificity to firefly luciferase (FLuc) and its derivatives such as AkaLuc. However, the brightness of Antares in mice is limited by the poor solubility and bioavailability of the NanoLuc substrate furimazine. Here, we report a new substrate, hydrofurimazine, whose enhanced aqueous solubility allows delivery of higher doses to mice. In the liver, Antares with hydrofurimazine exhibited similar brightness to AkaLuc with its substrate AkaLumine. Further chemical exploration generated a second substrate, fluorofurimazine, with even higher brightness in vivo. We used Antares with fluorofurimazine to track tumor size and AkaLuc with AkaLumine to visualize CAR-T cells within the same mice, demonstrating the ability to perform two-population imaging with these two luciferase systems.


Sujet(s)
Furanes/composition chimique , Luciferases/composition chimique , Mesures de luminescence/méthodes , Protéines luminescentes/composition chimique , Animaux , Dosages enzymatiques/méthodes , Spécificité du substrat
12.
J Med Chem ; 62(10): 4884-4901, 2019 05 23.
Article de Anglais | MEDLINE | ID: mdl-31013090

RÉSUMÉ

Novel pyridine- and pyrimidine-based allosteric inhibitors are reported that achieve PDE4D subtype selectivity through recognition of a single amino acid difference on a key regulatory domain, known as UCR2, that opens and closes over the catalytic site for cAMP hydrolysis. The design and optimization of lead compounds was based on iterative analysis of X-ray crystal structures combined with metabolite identification. Selectivity for the activated, dimeric form of PDE4D provided potent memory enhancing effects in a mouse model of novel object recognition with improved tolerability and reduced vascular toxicity over earlier PDE4 inhibitors that lack subtype selectivity. The lead compound, 28 (BPN14770), has entered midstage, human phase 2 clinical trials for the treatment of Fragile X Syndrome.


Sujet(s)
Encéphalopathies/traitement médicamenteux , Cyclic Nucleotide Phosphodiesterases, Type 4/métabolisme , Conception de médicament , Syndrome du chromosome X fragile/traitement médicamenteux , Inhibiteurs de la phosphodiestérase-4/synthèse chimique , Régulation allostérique/effets des médicaments et des substances chimiques , Animaux , Comportement animal/effets des médicaments et des substances chimiques , Encéphalopathies/enzymologie , Cyclic Nucleotide Phosphodiesterases, Type 4/génétique , Syndrome du chromosome X fragile/enzymologie , Humains , Concentration inhibitrice 50 , Mâle , Souris de lignée ICR , Structure moléculaire , Inhibiteurs de la phosphodiestérase-4/composition chimique , Inhibiteurs de la phosphodiestérase-4/pharmacologie , Relation structure-activité
13.
Org Biomol Chem ; 15(40): 8559-8567, 2017 Oct 18.
Article de Anglais | MEDLINE | ID: mdl-28972606

RÉSUMÉ

We report the synthesis and characterization of novel coelenterazine analogues that demonstrate a red-shift in their bioluminescent emission with NanoLuc luciferase. These coelenterazines can be tuned to shift the bioluminescent emission from blue light in the native system. In particular, direct attachment of an aryl moiety to the imidazopyrazinone core of furimazine at the C8 position provides a significant red-shift while maintaining reasonable light output. In addition, modification of the C6 aryl moiety provided additive red-shifts, and by combining the most promising modifications we report a coelenterazine with a maximum emission near 600 nm with NanoLuc. Finally, we show that this new bioluminescent system is capable of efficient BRET to far-red fluorophores. We anticipate these new principles of NanoLuc substrate design will impact applications that depend on shifting the colour of emission to the red, most notably in vivo bioluminescent imaging.


Sujet(s)
Imidazoles/composition chimique , Luciferases/composition chimique , Luminescents/composition chimique , Pyrazines/composition chimique , Imidazoles/métabolisme , Luciferases/métabolisme , Luminescents/métabolisme , Mesures de luminescence , Structure moléculaire , Pyrazines/métabolisme
14.
ACS Chem Biol ; 12(4): 1028-1037, 2017 04 21.
Article de Anglais | MEDLINE | ID: mdl-28195704

RÉSUMÉ

Novel engineered NanoLuc (Nluc) luciferase being smaller, brighter, and superior to traditional firefly (Fluc) or Renilla (Rluc) provides a great opportunity for the development of numerous biological, biomedical, clinical, and food and environmental safety applications. This new platform created an urgent need for Nluc inhibitors that could allow selective bioluminescent suppression and multiplexing compatibility with existing luminescence or fluorescence assays. Starting from thienopyrrole carboxylate 1, a hit from a 42 000 PubChem compound library with a low micromolar IC50 against Nluc, we derivatized four different structural fragments to discover a family of potent, single digit nanomolar, cell permeable inhibitors. Further elaboration revealed a channel that allowed access to the external Nluc surface, resulting in a series of highly potent cell impermeable Nluc inhibitors with negatively charged groups likely extending to the protein surface. The permeability was evaluated by comparing EC50 shifts calculated from both live and lysed cells expressing Nluc cytosolically. Luminescence imaging further confirmed that cell permeable compounds inhibit both intracellular and extracellular Nluc, whereas less permeable compounds differentially inhibit extracellular Nluc and Nluc on the cell surface. The compounds displayed little to no toxicity to cells and high luciferase specificity, showing no activity against firefly luciferase or even the closely related NanoBit system. Looking forward, the structural motifs used to gain access to the Nluc surface can also be appended with other functional groups, and therefore interesting opportunities for developing assays based on relief-of-inhibition can be envisioned.


Sujet(s)
Perméabilité des membranes cellulaires , Antienzymes/pharmacologie , Luciferases/antagonistes et inhibiteurs , Antienzymes/composition chimique , Antienzymes/métabolisme , Cellules HEK293 , Humains , Interactions hydrophobes et hydrophiles , Concentration inhibitrice 50 , Relation structure-activité
15.
Chemistry ; 22(30): 10369-75, 2016 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-27305599

RÉSUMÉ

The growing popularity of bioluminescent assays has highlighted the need for coelenterazine analogues possessing properties tuned for specific applications. However, the structural diversity of known coelenterazine analogues has been limited by current syntheses. Known routes for the preparation of coelenterazine analogues employ harsh reaction conditions that limit access to many substituents and functional groups. Novel synthetic routes reported here establish simple and robust methods for synthesis and investigation of structurally diverse marine luciferase substrates. Specifically, these new routes allow synthesis of coelenterazine analogues containing various heterocyclic motifs and substituted aromatic groups with diverse electronic substituents at the R(2) position. Interesting analogues described herein were characterized by their physicochemical properties, bioluminescent half-life, light output, polarity and cytotoxicity. Some of the analogues represent leads that can be utilized in the development of improved bioluminescent systems.

16.
J Carbohydr Chem ; 35(5): 249-260, 2016.
Article de Anglais | MEDLINE | ID: mdl-28626297

RÉSUMÉ

Improvements in the synthesis of carbon-linked glucuronide/glucoside conjugates of cancer chemopreventive retinoids have been achieved starting with 2,3,4,6-tetra-O-benzyl-D-glucopyranose. The revised approach demonstrates better yields, eliminates the use of an expensive, carcinogenic protecting group reagent, and avoids much painstaking chromatography. The new approach should allow synthesis of larger quantities of the agents for detailed animal and mechanistic studies.

17.
J Am Chem Soc ; 130(14): 4906-13, 2008 Apr 09.
Article de Anglais | MEDLINE | ID: mdl-18345677

RÉSUMÉ

Isopentenyl diphosphate isomerase (IDI) catalyzes the interconversion of isopentenyl diphosphate (IPP) and dimethylallyl diphosphate (DMAPP), the basic building blocks of isoprenoid molecules. Two structurally unrelated classes of IDI are known. Type I IPP isomerase (IDI-1) utilizes a divalent metal in a protonation-deprotonation reaction; whereas, the type II enzyme (IDI-2) requires reduced flavin. Epoxy, diene, and fluorinated substrate analogues, irreversible inhibitors of IDI-1, were analyzed as mechanistic probes for IDI-2. 3,4-Oxido-3-methyl-1-butyl diphosphate (eIPP), 3-methylene-4-penten-1-yl diphosphate (vIPP), and 3-(fluoromethyl)-3-buten-1-yl diphosphate (fmIPP) inactivate IDI-2 through formation of covalent adducts with the reduced flavin. UV-visible spectra of the inactivated complexes are consistent with modification of the isoalloxazine ring at position N5. vIPP and fmIPP are also alternate substrates with isomerization competing with alkylation of the flavin cofactor. (Z)-3-(Fluoromethyl)-2-buten-1-yl diphosphate ((Z)-fmDMAPP) and (Z)-3-(difluoromethyl)-2-buten-1-yl diphosphate ((Z)-dfmDMAPP) are alternate substrates, which are isomerized to the corresponding IPP derivatives. The rates of isomerization of fmIPP and (Z)-fmDMAPP are approximately 50-fold less than IPP and DMAPP, respectively. dfmIPP is not an irreversible inhibitor. These studies indicate that the irreversible inhibitors inactivate the reduced flavin required for catalysis by electrophilic alkylation and are consistent with a protonation-deprotonation mechanism for the isomerization catalyzed by IDI-2.


Sujet(s)
Carbon-carbon double bond isomerases/métabolisme , Flavine mononucléotide/métabolisme , Fixation compétitive , Carbon-carbon double bond isomerases/antagonistes et inhibiteurs , Carbon-carbon double bond isomerases/composition chimique , Activation enzymatique , Flavine mononucléotide/composition chimique , Chromatographie gazeuse-spectrométrie de masse , Hémiterpènes , Isoenzymes , Cinétique , Spectrophotométrie UV , Thermus thermophilus/enzymologie
18.
J Org Chem ; 73(2): 726-9, 2008 Jan 18.
Article de Anglais | MEDLINE | ID: mdl-18088143

RÉSUMÉ

Type 2 isopentenyl diphosphate isomerase (IDI-2), which catalyzes the interconversion of isopentenyl diphosphate and dimethylallyl diphosphate, contains a tightly bound molecule of FMN. To probe the mechanism of the reaction, cyclopropyl and epoxy substrate analogues, designed to be mechanism-based irreversible inhibitors, were synthesized and evaluated with IDI-2 from Thermus thermophilus. The cyclopropyl analogues were alternative substrates. The epoxy analogue was an irreversible inhibitor, with kI = 0.37 +/- 0.07 min(-1) and KI = 1.4 +/- 0.3 microM. LC-MS studies revealed formation of an epoxide-FMN adduct.


Sujet(s)
Carbon-carbon double bond isomerases/antagonistes et inhibiteurs , Cyclopropanes/synthèse chimique , Cyclopropanes/pharmacologie , Composés époxy/synthèse chimique , Composés époxy/pharmacologie , Sites de fixation/effets des médicaments et des substances chimiques , Carbon-carbon double bond isomerases/composition chimique , Cyclopropanes/composition chimique , Évaluation préclinique de médicament , Activation enzymatique/effets des médicaments et des substances chimiques , Composés époxy/composition chimique , Hémiterpènes , Structure moléculaire , Structure tertiaire des protéines , Protéines recombinantes/antagonistes et inhibiteurs , Protéines recombinantes/composition chimique , Stéréoisomérie , Relation structure-activité
20.
Bioorg Med Chem ; 14(9): 3038-48, 2006 May 01.
Article de Anglais | MEDLINE | ID: mdl-16412653

RÉSUMÉ

All-trans retinoic acid analogues such as N-(4-hydroxyphenyl)retinamide (4-HPR) are effective chemopreventive and chemotherapeutic agents but their utility has been hampered by dose-limiting side effects. The glucuronide derivatives of 4-HPR, the oxygen-linked 4-HPROG and the carbon-linked 4-HPRCG, have been found to be more effective agents. The synthetic route to the fully C-linked analogue of 4-HPROG (4-HBRCG), which employs Suzuki coupling and Umpolung chemistries as key methodologies, is shown. The results of this study show 4-HBRCG to be an effective chemotherapeutic agent in a rat mammary tumor model while being devoid of classical retinoid toxicities.


Sujet(s)
Fenrétinide/composition chimique , Fenrétinide/usage thérapeutique , Glucuronides/composition chimique , 7,12-Diméthyl-benzo[a]anthracène/pharmacologie , Animaux , Tumeurs du sein/induit chimiquement , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Femelle , Fenrétinide/synthèse chimique , Fenrétinide/pharmacocinétique , Structure moléculaire , Rats , Rat Sprague-Dawley
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...