Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 322
Filter
1.
Chemistry ; 30(27): e202400272, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38445549

ABSTRACT

Springtails use unique compounds for their outermost epicuticular wax layer, often of terpenoid origin. We report here the structure and synthesis of socialane, the major cuticular constituent of the Collembola Hypogastrura socialis. Socialane is also the first regular nonaprenyl terpene with a cyclic head group. The saturated side chain has seven stereogenic centers, making the determination of the configuration difficult. We describe here the identification of socialane and a synthetic approach using the building blocks farnesol and phytol, enantioselective hydrogenation, and α-alkylation of sulfones for the synthesis of various stereoisomers. NMR experiments showed the presence of an anti-configuration of the methyl groups closest to the benzene ring and that the other methyl groups of the polyprenyl side-chain are not uniformly configured. Furthermore, socialane is structurally different from [6+2]-terpene viaticene of the closely related H. viatica, showing species specificity of the epicuticular lipids of this genus and hinting at a possible role of surface lipids in the communication of these gregarious arthropods.


Subject(s)
Arthropods , Terpenes , Animals , Stereoisomerism , Terpenes/chemistry , Arthropods/chemistry , Lipids/chemistry , Farnesol/chemistry , Farnesol/analogs & derivatives , Phytol/chemistry , Magnetic Resonance Spectroscopy , Hydrogenation
3.
Acta Pharmacol Sin ; 43(7): 1843-1856, 2022 Jul.
Article in English | MEDLINE | ID: mdl-34845369

ABSTRACT

Ras has long been viewed as a promising target for cancer therapy. Farnesylthiosalicylic acid (FTS), as the only Ras inhibitor has ever entered phase II clinical trials, has yielded disappointing results due to its strong hydrophobicity, poor tumor-targeting capacity, and low therapeutic efficiency. Thus, enhancing hydrophilicity and tumor-targeting capacity of FTS for improving its therapeutic efficacy is of great significance. In this study we conjugated FTS with a cancer-targeting small molecule dye IR783 and characterized the anticancer properties of the conjugate FTS-IR783. We showed that IR783 conjugation greatly improved the hydrophilicity, tumor-targeting and therapeutic potential of FTS. After a single oral administration in Balb/c mice, the relative bioavailability of FTS-IR783 was increased by 90.7% compared with FTS. We demonstrated that organic anion transporting polypeptide (OATP) and endocytosis synergistically drove the uptake of the FTS-IR783 conjugate in breast cancer MDA-MB-231 cells, resulting in superior tumor-targeting ability of the conjugate both in vitro and in vivo. We further revealed that FTS-IR783 conjugate could bind with and directly activate AMPK rather than affecting Ras, and subsequently regulate the TSC2/mTOR signaling pathway, thus achieving 2-10-fold increased anti-cancer therapeutic efficacy against 6 human breast cancer cell lines compared to FTS both in vivo and in vitro. Overall, our data highlights a promising approach for the modification of the anti-tumor drug FTS using IR783 and makes it possible to return FTS back to the clinic with a better efficacy.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Farnesol/analogs & derivatives , Farnesol/pharmacology , Farnesol/therapeutic use , Female , Humans , Mice , Salicylates , ras Proteins/metabolism , ras Proteins/therapeutic use
4.
Int J Nanomedicine ; 16: 6441-6453, 2021.
Article in English | MEDLINE | ID: mdl-34584410

ABSTRACT

BACKGROUND: Renal fibrosis is the common pathway in chronic kidney diseases progression to end-stage renal disease, but to date, no clinical drug for its treatment is approved. It has been demonstrated that the inhibitor of proto-oncogene Ras, farnesylthiosalicylic acid (FTS), shows therapeutic potential for renal fibrosis, but its application was hindered by the water-insolubility and low bioavailability. Hence, in this study, we improved these properties of FTS by encapsulating it into bovine serum albumin nanoparticles (AN-FTS) and tested its therapeutic effect in renal fibrosis. METHODS: AN-FTS was developed using a classic emulsification-solvent ultrasonication. The pharmacokinetics of DiD-loaded albumin nanoparticle were investigated in SD rats. The biodistribution and therapeutic efficacy of AN-FTS was assessed in a mouse model of renal fibrosis induced by unilateral ureteral obstruction (UUO). RESULTS: AN-FTS showed a uniform spherical shape with the size of 100.6 ± 1.12 nm and PDI < 0.25. In vitro, AN-FTS displayed stronger inhibitory effects on the activation of renal fibroblasts cells NRK-49F than free FTS. In vivo, AN-FTS showed significantly higher peak concentration and area under the concentration-time curve. After intravenous administration to UUO-induced renal fibrosis mice, AN-FTS accumulated preferentially in the fibrotic kidney, and alleviated renal fibrosis and inflammation significantly more than the free drug. Mechanistically, the improved anti-fibrosis effect of AN-FTS was associated with greater inhibition in renal epithelial-to-mesenchymal transformation process via Ras/Raf1/p38 signaling pathway. CONCLUSION: The study reveals that AN-FTS is capable of delivering FTS to fibrotic kidney and showed superior therapeutic efficacy for renal fibrosis.


Subject(s)
Nanoparticles , Signal Transduction , Albumins , Animals , Farnesol/analogs & derivatives , Fibrosis , Mice , Proto-Oncogene Proteins c-raf , Rats , Rats, Sprague-Dawley , Salicylates , Tissue Distribution
5.
J Nanobiotechnology ; 19(1): 177, 2021 Jun 12.
Article in English | MEDLINE | ID: mdl-34118951

ABSTRACT

BACKGROUND: Ras activation is a frequent event in hepatocellular carcinoma (HCC). Combining a RAS inhibitor with traditional clinical therapeutics might be hampered by a variety of side effects, thus hindering further clinical translation. Herein, we report on integrating an IR820 nanocapsule-augmented sonodynamic therapy (SDT) with the RAS inhibitor farnesyl-thiosalicylic acid (FTS). Using cellular and tumor models, we demonstrate that combined nanocapsule-augmented SDT with FTS induces an anti-tumor effect, which not only inhibits tumor progression, and enables fluorescence imaging. To dissect the mechanism of a combined tumoricidal therapeutic strategy, we investigated the scRNA-seq transcriptional profiles of an HCC xenograft following treatment. RESULTS: Integrative single-cell analysis identified several clusters that defined many corresponding differentially expressed genes, which provided a global view of cellular heterogeneity in HCC after combined SDT/FTS treatment. We conclude that the combination treatment suppressed HCC, and did so by inhibiting endothelial cells and a modulated immunity. Moreover, hepatic stellate secretes hepatocyte growth factor, which plays a key role in treating SDT combined FTS. By contrast, enrichment analysis estimated the functional roles of differentially expressed genes. The Gene Ontology terms "cadherin binding" and "cell adhesion molecule binding" and KEGG pathway "pathway in cancer" were significantly enriched by differentially expressed genes after combined SDT/FTS therapy. CONCLUSIONS: Thus, some undefined mechanisms were revealed by scRNA-seq analysis. This report provides a novel proof-of-concept for combinatorial HCC-targeted therapeutics that is based on a non-invasive anti-tumor therapeutic strategy and a RAS inhibitor.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Diathermy/methods , Liver Neoplasms/drug therapy , Sequence Analysis, RNA , ras Proteins/antagonists & inhibitors , Animals , Carcinoma, Hepatocellular/radiotherapy , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Endothelial Cells , Farnesol/analogs & derivatives , Farnesol/pharmacology , Female , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Liver Neoplasms/radiotherapy , Mice, Inbred BALB C , Mice, Nude , Salicylates
6.
Biosci Biotechnol Biochem ; 85(7): 1628-1638, 2021 Jun 24.
Article in English | MEDLINE | ID: mdl-33890631

ABSTRACT

Juvenile hormone III (JH III) plays an important role in insect reproduction, development, and behavior. The second branch of JH III production includes oxidation of farnesol to farnesal by farnesol dehydrogenase. This study reported the identification and characterization of Plutella xylostella farnesol dehydrogenase (PxFoLDH). Our results showed that PxFoLDH belongs to the short-chain dehydrogenase/reductase superfamily, consisting of a single domain with a structurally conserved Rossman fold, an NAD(P) (H)-binding region and a structurally diverse C-terminal region. The purified enzyme displayed maximum activity at 55$\ $°C with pH 9.5 and was stable in the temperature below 70$\ ^\circ $C. PxFoLDH was determined to be a monomer with a relative molecular weight of 27 kDa and highly specific for trans, trans-farnesol, and NADP+. Among analog inhibitors tested, farnesyl acetate was the most effective inhibitor with the lowest Ki value of 0.02 µm. Our findings showed this purified enzyme may represent as NADP+-farnesol dehydrogenase.


Subject(s)
Insecticides/pharmacology , Lepidoptera/enzymology , NAD (+) and NADP (+) Dependent Alcohol Oxidoreductases/antagonists & inhibitors , NADP/chemistry , Animals , Enzyme Inhibitors/pharmacology , Farnesol/analogs & derivatives , Farnesol/pharmacology , Hydrogen-Ion Concentration , Insecticides/chemistry , Kinetics , NAD (+) and NADP (+) Dependent Alcohol Oxidoreductases/chemistry , NAD (+) and NADP (+) Dependent Alcohol Oxidoreductases/metabolism , Substrate Specificity , Temperature
7.
BMC Cancer ; 21(1): 237, 2021 Mar 06.
Article in English | MEDLINE | ID: mdl-33676427

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC), one of the most lethal cancers, is driven by oncogenic KRAS mutations. Farnesyl thiosalicylic acid (FTS), also known as salirasib, is a RAS inhibitor that selectively dislodges active RAS proteins from cell membrane, inhibiting downstream signaling. FTS has demonstrated limited therapeutic efficacy in PDAC patients despite being well tolerated. METHODS: To improve the efficacy of FTS in PDAC, we performed a genome-wide CRISPR synthetic lethality screen to identify genetic targets that synergize with FTS treatment. Among the top candidates, multiple genes in the endoplasmic reticulum-associated protein degradation (ERAD) pathway were identified. The role of ERAD inhibition in enhancing the therapeutic efficacy of FTS was further investigated in pancreatic cancer cells using pharmaceutical and genetic approaches. RESULTS: In murine and human PDAC cells, FTS induced unfolded protein response (UPR), which was further augmented upon treatment with a chemical inhibitor of ERAD, Eeyarestatin I (EerI). Combined treatment with FTS and EerI significantly upregulated the expression of UPR marker genes and induced apoptosis in pancreatic cancer cells. Furthermore, CRISPR-based genetic ablation of the key ERAD components, HRD1 and SEL1L, sensitized PDAC cells to FTS treatment. CONCLUSION: Our study reveals a critical role for ERAD in therapeutic response of FTS and points to the modulation of UPR as a novel approach to improve the efficacy of FTS in PDAC treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Endoplasmic Reticulum-Associated Degradation/drug effects , Pancreatic Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , CRISPR-Cas Systems/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Endoplasmic Reticulum-Associated Degradation/genetics , Farnesol/analogs & derivatives , Farnesol/pharmacology , Farnesol/therapeutic use , Gene Knockout Techniques , Humans , Hydrazones/pharmacology , Hydrazones/therapeutic use , Hydroxyurea/analogs & derivatives , Hydroxyurea/pharmacology , Hydroxyurea/therapeutic use , Mice , Pancreatic Neoplasms/pathology , Proteins/genetics , Salicylates/pharmacology , Salicylates/therapeutic use , Synthetic Lethal Mutations , Ubiquitin-Protein Ligases/genetics , Unfolded Protein Response/drug effects
9.
Int J Med Sci ; 17(12): 1795-1802, 2020.
Article in English | MEDLINE | ID: mdl-32714082

ABSTRACT

Background/Aim: Pancreatic adenocarcinoma is a highly malignant tumor. Synergistic combinations of anticancer agents for the effective treatment of pancreatic cancer patients are urgently needed. Here, we investigated the combined effect of celecoxib (CEL) and salirasib (SAL) on pancreatic cancer cells. Methods: Cell viability and apoptosis were measured by the trypan blue assay, three-dimensional cultures, propidium iodide staining, and caspase-3 assay. NF-κB activation and the protein levels of Akt, pAkt, and Bcl-2 were determined by the luciferase reporter assay and western blot. Results: Co-treatment with CEL and SAL had stronger effects on decreasing cell viability and inducing apoptosis in Panc-1 cells as compared with each agent individually. This combination strongly inhibited NF-κB activity and reduced pAkt and Bcl-2 levels in Panc-1 cells. Conclusion: SAL in combination with CEL may represent a new approach for effective inhibition of pancreatic cancer.


Subject(s)
Celecoxib/pharmacology , Farnesol/analogs & derivatives , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Salicylates/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Caspase 3/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Farnesol/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , NF-kappa B/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Signal Transduction
10.
J Nanobiotechnology ; 18(1): 89, 2020 Jun 11.
Article in English | MEDLINE | ID: mdl-32527262

ABSTRACT

BACKGROUND: Farnesol is a sesquiterpene from propolis and citrus fruit that shows promising anti-bacterial activity for caries treatment and prevention, but its hydrophobicity limits the clinical application. We aimed to develop the novel polymeric micelles (PMs) containing a kind of derivative of farnesol and a ligand of pyrophosphate (PPi) that mediated PMs to adhere tightly with the tooth enamel. RESULTS: Farnesal (Far) was derived from farnesol and successfully linked to PEG via an acid-labile hydrazone bond to form PEG-hyd-Far, which was then conjugated to PPi and loaded into PMs to form the aimed novel drug delivery system, PPi-Far-PMs. The in vitro test about the binding of PPi-Far-PMs to hydroxyapatite showed that PPi-Far-PMs could bind rapidly to hydroxyapatite and quickly release Far under the acidic conditions. Results from the mechanical testing and the micro-computed tomography indicated that PPi-Far-PMs could restore the microarchitecture of teeth with caries. Moreover, PPi-Far-PMs diminished the incidence and severity of smooth and sulcal surface caries in rats that were infected with Streptococcus mutans while being fed with a high-sucrose diet. The anti-caries efficacy of free Far can be improved significantly by PPi-Far-PMs through the effective binding of it with tooth enamel via PPi. CONCLUSIONS: This novel drug-delivery system may be useful for the treatment and prevention of dental caries as well as the targeting therapy of anti-bacterial drugs in the oral disease.


Subject(s)
Cariostatic Agents , Dental Caries , Durapatite , Farnesol/analogs & derivatives , Micelles , Animals , Cariostatic Agents/chemistry , Cariostatic Agents/pharmacokinetics , Cariostatic Agents/pharmacology , Dental Caries/drug therapy , Dental Caries/metabolism , Dental Caries/pathology , Diphosphates/chemistry , Diphosphates/pharmacokinetics , Diphosphates/pharmacology , Drug Carriers , Durapatite/chemistry , Durapatite/metabolism , Farnesol/chemistry , Farnesol/pharmacokinetics , Farnesol/pharmacology , Hydrophobic and Hydrophilic Interactions , Molar/drug effects , Molar/ultrastructure , Polyethylene Glycols/chemistry , Rats , Streptococcus mutans/drug effects
11.
Int J Mol Sci ; 21(10)2020 May 12.
Article in English | MEDLINE | ID: mdl-32408605

ABSTRACT

Glia cells are involved in upper motor neuron degeneration in amyotrophic lateral sclerosis (ALS). Protease activated receptor 1 (PAR1) pathway is related to brain pathologies. Brain PAR1 is located on peri-synaptic astrocytes, adjacent to pyramidal motor neurons, suggesting possible involvement in ALS. Brain thrombin activity in superoxide dismutase 1 (SOD1) mice was measured using a fluorometric assay, and PAR1 levels by western blot. PAR1 was localized using immunohistochemistry staining. Treatment targeted PAR1 pathway on three levels; thrombin inhibitor TLCK (N-Tosyl-Lys-chloromethylketone), PAR1 antagonist SCH-79797 and the Ras intracellular inhibitor FTS (S-trans-trans-farnesylthiosalicylic acid). Mice were weighed and assessed for motor function and survival. SOD1 brain thrombin activity was increased (p < 0.001) particularly in the posterior frontal lobe (p = 0.027) and hindbrain (p < 0.01). PAR1 levels were decreased (p < 0.001, brain, spinal cord, p < 0.05). PAR1 and glial fibrillary acidic protein (GFAP) staining decreased in the cerebellum and cortex. SOD1 mice lost weight (≥17 weeks, p = 0.047), and showed shorter rotarod time (≥14 weeks, p < 0.01). FTS 40mg/kg significantly improved rotarod scores (p < 0.001). Survival improved with all treatments (p < 0.01 for all treatments). PAR1 antagonism was the most efficient, with a median survival improvement of 10 days (p < 0.0001). Our results support PAR1 pathway involvement in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Brain/metabolism , Disease Models, Animal , Receptor, PAR-1/metabolism , Superoxide Dismutase-1/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Astrocytes/metabolism , Body Weight/drug effects , Farnesol/analogs & derivatives , Farnesol/pharmacology , Glial Fibrillary Acidic Protein/metabolism , Humans , Mice , Mice, Transgenic , Motor Neurons/metabolism , Mutation , Pyrroles/pharmacology , Quinazolines/pharmacology , Salicylates/pharmacology , Signal Transduction/drug effects , Superoxide Dismutase-1/genetics , Survival Analysis , Tosyllysine Chloromethyl Ketone/pharmacology
12.
Drug Des Devel Ther ; 13: 4053-4063, 2019.
Article in English | MEDLINE | ID: mdl-31819374

ABSTRACT

BACKGROUND: Alzheimer's disease (AD), a leading cause of dementia, becomes a serious health issue for individuals and society around the world. AD is a neurodegenerative disease characterized by the deposition of amyloid-ß (Aß) peptides and neurofibrillary tangles (NFT) and the loss of large numbers of neurons. To date, there is no effective treatment for AD, and thus, to enhance neurogenesis in the AD brain may be a therapeutic strategy. RAS signaling pathway involves in synaptic plasticity and memory formation, which is overexpressed in brains with AD. This study used Aß1-42-injected mice (Aß1-42-mice) as the AD model to investigate the effects of S-trans, trans-farnesylthiosalicylic acid (FTS), a synthetic Ras inhibitor, on the impairment of neurogenesis and the spatial cognitive deficits. MATERIALS AND METHODS: AD model mice were manufactured through intracerebroventricular injection of Aß1-42. Morris water maze (MWM) was performed to evaluate the capacity of spatial memory, and Nissl staining was applied to assess neuronal damage in the hippocampus CA1. Immunohistochemistry of 5-bromo-2-deoxyuridine (BrdU), BrdU/neuronal nuclei (NeuN), and doublecortin (DCX) were used to detect progenitor cell proliferation, maturation, and neurite growth, respectively. And the expression levels of RAS, ERK/ERK phosphorylation (p-ERK) and CREB/CREB phosphorylation (p-CREB) were detected by Western blot. RESULTS: The results demonstrated that FTS could prevent Aß1-42 to impair survival and neurite growth of newborn neurons in the hippocampal dentate gyrus (DG) in Aß1-42-mice. Furthermore, behavioral indexes and morphological findings showed that FTS improved the learning and spatial memory abilities of Aß1-42-mice. In addition, FTS could inhibit the levels of hippocampal p-ERK and p-CREB activated by Aß, which is the underlying molecular mechanism. CONCLUSION: In conclusion, these findings suggest that FTS as a RAS inhibitor could be a potential therapeutic agent for the treatment of AD.


Subject(s)
Alzheimer Disease/drug therapy , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Farnesol/analogs & derivatives , MAP Kinase Signaling System/drug effects , Neuroprotective Agents/pharmacology , Salicylates/pharmacology , ras Proteins/antagonists & inhibitors , Alzheimer Disease/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Doublecortin Protein , Extracellular Signal-Regulated MAP Kinases/metabolism , Farnesol/administration & dosage , Farnesol/chemistry , Farnesol/pharmacology , Injections, Intraperitoneal , Male , Maze Learning/drug effects , Mice , Mice, Inbred ICR , Molecular Structure , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemistry , Salicylates/administration & dosage , Salicylates/chemistry , Stereoisomerism , ras Proteins/metabolism
13.
PLoS One ; 14(8): e0220718, 2019.
Article in English | MEDLINE | ID: mdl-31430298

ABSTRACT

A series of experiments were conducted to characterize a novel restorative material. We explored the effect on biological, physical and chemical properties of glass ionomer cement (GIC) adding-the naturally occurring tt-farnesol (900 mM). Two groups were accomplished for all assays: GIC+tt-farnesol and GIC (control). Biological assays: 1) agar diffusion against some cariogenic bacteria; 2) S. mutans biofilm formation and confocal laser scanning microscopy-CLSM. 3) gtfB, gtfC, gtfD, gbpB, vicR, and covR expression; 4) MTT and microscopic morphology. Physical properties assays: 1) roughness; 2) hardness; 3) compressive strength and 4) diametral tensile strength. Chemical assay: Raman spectroscopy. The adding of tt-farnesol to GIC led to larger zones of inhibition (p<0.05), biofilms with a short-term reduction in bacterial viability but similar biomass (p>0.05). Polysaccharides levels increased over time, similarly over groups (p>0.05). Viable and non-viable S. mutans were seen on the specimens' surface by CLSM but their virulence was not modulated by tt-farnesol. The tt-farnesol increased the HaCaT cell viability without impact on compressive and diametral tensile strength and roughness although the hardness was positively affected (p<0.05). Raman confirmed the presence of tt-farnesol. The incorporation of tt-farnesol into GIC inhibited the growth of cariogenic bacteria but had a little effect on the composition, structure and physiology of the biofilm matrices. Also, the tt-farnesol increased the hardness and the biocompatibility of the GIC, not influencing negatively other physical properties of the restorative material.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Farnesol/analogs & derivatives , Farnesol/pharmacology , Glass Ionomer Cements/chemistry , Glass Ionomer Cements/pharmacology , Biofilms/drug effects , Biofilms/growth & development , Hardness , Humans , Materials Testing , Microbial Viability/drug effects , Streptococcus mutans/drug effects , Streptococcus mutans/physiology , Tensile Strength
14.
J Chem Ecol ; 45(8): 667-672, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31313134

ABSTRACT

Agriotes ustulatus is an economically important click beetle in Europe. A female-produced pheromone, (E,E)-farnesyl acetate, has been identified and is used for monitoring and detecting males. More recently, a floral lure targeting females with modest, but significant, activity has been described. Based on preliminary data, we hypothesized, that similar to the effects on the congeneric A. brevis, addition of the pheromone to the floral lure should improve female A. ustulatus catches. Also, as click beetles have been reported to respond to white light, we studied possible interactions between visual and chemical cues. In field trials, the addition of the synthetic pheromone to the floral lure resulted in a dramatic increase in the number of females trapped, whereas male catches remained unaffected and equal to those in traps baited with pheromone only. A white visual cue did not influence trap catches. Maximum catches of both sexes of A. ustulatus can be achieved using the pheromone and the floral lure inside the same trap. Furthermore, the compounds can be formulated in a single polyethylene bag dispenser, making handling of the trap easier. Due to a much larger proportion of females in the catch, this improved trap may be a promising tool for semiochemical-based, environmentally sound agricultural practice against this important pest.


Subject(s)
Coleoptera/physiology , Pheromones/chemistry , Acrolein/analogs & derivatives , Acrolein/chemistry , Acrolein/pharmacology , Allylbenzene Derivatives , Animals , Anisoles/chemistry , Anisoles/pharmacology , Behavior, Animal/drug effects , Farnesol/analogs & derivatives , Farnesol/chemistry , Farnesol/pharmacology , Female , Flowers/chemistry , Flowers/metabolism , Insect Control , Male , Pheromones/pharmacology , Stereoisomerism
15.
Fish Shellfish Immunol ; 88: 150-160, 2019 May.
Article in English | MEDLINE | ID: mdl-30794934

ABSTRACT

The white Spot Syndrome Virus (WSSV) is a pathogen that causes huge economic losses in the shrimp-farming industry globally. At the WSSV genome replication stage (12 hpi) in WSSV-infected shrimp hemocytes, activation of the PI3K-Akt-mTOR pathway triggers metabolic changes that resemble the Warburg effect. In shrimp, the upstream regulators of this pathway are still unknown, and in the present study, we isolate, characterize and investigate two candidate factors, i.e. the shrimp Ras GTPase isoforms LvRas and LvRap, both of which are upregulated after WSSV infection. dsRNA silencing experiments show that virus replication is significantly reduced when expression of either of these genes is suppressed. Pretreatment with the Ras inhibitor Salirasib further suggests that LvRas, which is a homolog to a commonly overexpressed human oncoprotein, may be involved in regulating the WSSV-induced Warburg effect. We also show that while both the PI3K-Akt-mTOR and Raf-MEK-ERK pathways are activated by WSSV infection, LvRas appears to be involved only in the regulation of the mTOR pathway.


Subject(s)
Penaeidae/virology , Virus Replication/genetics , White spot syndrome virus 1/genetics , rab GTP-Binding Proteins/metabolism , ras Proteins/metabolism , Animals , Aquaculture , Enzyme Inhibitors/pharmacology , Farnesol/analogs & derivatives , Farnesol/pharmacology , Hemocytes/virology , Penaeidae/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Double-Stranded/genetics , Salicylates/pharmacology , TOR Serine-Threonine Kinases/metabolism , White spot syndrome virus 1/metabolism , rab GTP-Binding Proteins/genetics , ras Proteins/genetics
16.
Int Immunopharmacol ; 68: 218-225, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30658315

ABSTRACT

Inducible nitric oxide synthase (iNOS) is a molecule critical for the development of inflammation-associated disorders. Its induction should be tightly controlled in order to maintain cellular homeostasis. Upon lipopolysaccharide (LPS) stimulation, iNOS, in most settings, is induced by the activation of inhibitor of κB-α (IκB-α)-nuclear factor κB (NF-κB) signaling. Farnesyl thiosalicylic acid (FTS), a synthetic small molecule that is considered to detach Ras from the inner cell membrane, has been shown to exhibit numerous anti-inflammatory functions. However, it remains unclear whether and how it affects iNOS induction in macrophages. The present study addressed this issue in cultured macrophages and endotoxemic mice. Results showed that FTS pretreatment significantly prevented LPS-induced increases in iNOS protein and mRNA expression levels in murine cultured macrophages, which were confirmed in organs in vivo from endotoxemic mice, such as the liver and lung. Mechanistic studies revealed that FTS pretreatment did not affect IκB-α degradation and NF-κB activation in LPS-treated macrophages. The nuclear transport of the active NF-κB was also not affected by FTS. But FTS pretreatment reduced the binding of NF-κB to its DNA elements, and reduced NF-κB bindings to iNOS promoter inside LPS-treated macrophages. Finally, our results showed that FST pretreatment increased mouse survival rate compared to LPS alone treatment. Taken together, these results indicate that FTS attenuates iNOS induction in macrophages likely through inhibition of iNOS mRNA transcription, providing further insight into the molecular mechanism of action of FTS in inflammatory disorder therapy.


Subject(s)
Farnesol/analogs & derivatives , Macrophages/drug effects , Nitric Oxide Synthase Type II/genetics , Salicylates/pharmacology , Animals , Cells, Cultured , Farnesol/pharmacology , Lipopolysaccharides , Liver/drug effects , Liver/metabolism , Lung/drug effects , Lung/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , NF-kappa B/metabolism , Nitric Oxide/metabolism , RNA, Messenger/metabolism
17.
Molecules ; 23(11)2018 Nov 21.
Article in English | MEDLINE | ID: mdl-30469331

ABSTRACT

Hop-derived compounds have been subjected to numerous biomedical studies investigating their impact on a wide range of pathologies. Isomerised bitter acids (isoadhumulone, isocohumulone and isohumulone) from hops, used in the brewing process of beer, are known to inhibit members of the aldo-keto-reductase superfamily. Aldo-keto-reductase 1B10 (AKR1B10) is upregulated in various types of cancer and has been reported to promote carcinogenesis. Inhibition of AKR1B10 appears to be an attractive means to specifically treat RAS-dependent malignancies. However, the closely related reductases AKR1A1 and AKR1B1, which fulfil important roles in the detoxification of endogenous and xenobiotic carbonyl compounds oftentimes crossreact with inhibitors designed to target AKR1B10. Accordingly, there is an ongoing search for selective AKR1B10 inhibitors that do not interact with endogeneous AKR1A1 and AKR1B1-driven detoxification systems. In this study, unisomerised α-acids (adhumulone, cohumulone and n-humulone) were separated and tested for their inhibitory potential on AKR1A1, AKR1B1 and AKR1B10. Also AKR1B10-mediated farnesal reduction was effectively inhibited by α-acid congeners with Ki-values ranging from 16.79 ± 1.33 µM (adhumulone) to 3.94 ± 0.33 µM (n-humulone). Overall, α-acids showed a strong inhibition with selectivity (115⁻137 fold) for AKR1B10. The results presented herein characterise hop-derived α-acids as a promising basis for the development of novel and selective AKR1B10-inhibitors.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Cyclohexanones/pharmacology , Cyclohexenes/pharmacology , Terpenes/pharmacology , Aldehyde Reductase/metabolism , Aldo-Keto Reductases , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor , Farnesol/analogs & derivatives , Farnesol/chemistry , Gene Expression Regulation, Enzymologic/drug effects , Humans , Humulus/chemistry
18.
Bioresour Technol ; 269: 577-580, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30181019

ABSTRACT

Diminishing petroleum reserves and the rapid accumulation of greenhouse gases lead to increasing interest in microbial biofuels. In this study, a heterologous farnesyl acetate biosynthesis pathway was constructed in Escherichia coli for the first time. Firstly, the AtoB, ERG13, tHMG1, ERG12, ERG8, MVD1, Idi, IspA and PgpB were expressed to accumulate farnesol in the E. coli cells. Then the alcohol acetyltransferase (ATF1) was heterologous overexpressed for the subsequent esterification farnesol to farnesyl acetate. The engineered strain DG 106 accumulated 128 ±â€¯10.5 mg/L of farnesyl acetate. Finally, the isopentenyl-diphosphate isomerase was further overexpressed, and the recombinant strain DG107 produced 201 ±â€¯11.7 mg/L of farnesyl acetate. This study shows the novel method for the biosynthesis of the advanced biofuel farnesyl acetate directly from glucose and highlight the enormous designing strategies for metabolic engineering of bioproducts.


Subject(s)
Biofuels , Escherichia coli , Farnesol/analogs & derivatives , Metabolic Engineering , Escherichia coli Proteins , Phosphatidate Phosphatase
19.
Cancer Chemother Pharmacol ; 82(3): 511-519, 2018 09.
Article in English | MEDLINE | ID: mdl-29992354

ABSTRACT

PURPOSE: Patients with RAS-positive tumors respond poorly to chemotherapies and have a few treatment options. Salirasib is an oral RAS inhibitor that competitively blocks the membrane association of RAS proteins. The aim of this phase I multiple-ascending-dose clinical trial was to investigate the safety and pharmacokinetics of Salirasib in Japanese patients with relapsed/refractory solid tumors and to explore its efficacy. METHODS: Salirasib was started at a dose of 100-mg twice-daily and escalated to a maximum of 1000-mg twice-daily from days 1 to 21 of a 28-day regimen. The pharmacokinetics was evaluated on days 1 and 21. Dose-limiting toxicity (DLT) and adverse events (AEs) were monitored throughout the trial. Patients with stable disease or better repeated the dosing regimen. RESULTS: A total of 21 patients received Salirasib. Among 14 patients tested, 4 had KRAS mutations. Cmax and AUCinf were maximal at 800 mg. No maximum tolerable dose was discerned, as no DLT was observed in any dosing group. The most frequently observed AEs were gastrointestinal disturbances, including diarrhea, abdominal pain, and nausea. No AEs led to discontinuation. All patients completed the first regimen and 11 patients repeated the regimen (median: 2 cycles; range: 1-13). Patients with KRAS mutations showed median progression-free survival of 227 days (range: 79-373). CONCLUSION: Salirasib was safe and well tolerated in Japanese patients, and 800-mg twice-daily is recommended for phase II trials. Although the number of participants with KRAS mutations was limited, the remarkably long progression-free period warrants further investigation. CLINICAL TRIAL REGISTRATION: JAPIC Clinical Trials Information; JapicCTI-121751.


Subject(s)
Farnesol/analogs & derivatives , Neoplasms/drug therapy , Salicylates/administration & dosage , ras Proteins/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacokinetics , Farnesol/administration & dosage , Farnesol/adverse effects , Farnesol/pharmacokinetics , Female , Humans , Male , Middle Aged , Neoplasm Staging , Neoplasms/metabolism , Neoplasms/pathology , Salicylates/adverse effects , Salicylates/pharmacokinetics
20.
Int J Oncol ; 53(2): 725-736, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29901113

ABSTRACT

The active form of the small GTPase RAS binds to downstream effectors to promote cell growth and proliferation. RAS signal enhancement contributes to tumorigenesis, invasion, and metastasis in various different cancers. HRAS proto-oncogene GTPase (HRAS), one of the RAS isoforms, was the first human oncogene for which mutations were reported in T24 bladder cancer (BC) cells in 1982, and HRAS mutation or upregulation has been reported in several cancers. According to data from The Cancer Genome Atlas, HRAS expression was significantly upregulated in clinical BC samples compared to healthy samples (P=0.0024). HRAS expression was also significantly upregulated in BC with HRAS mutation compared to patients without HRAS mutation (P<0.0001). The tumor suppressive effect of salirasib, a RAS inhibitor, has been reported in several cancer types, but only at relatively high concentrations. As such, RAS inhibitors have not been used for clinical applications. The aim of the current study was to investigate the therapeutic potential of targeting HRAS using salirasib and small interfering RNA (siRNA) and to characterize the mechanism by which HRAS functions using recently developed quantitative in vitro proteome-assisted multiple reaction monitoring for protein absolute quantification (iMPAQT), in BC cells. iMPAQT allows measurement of the absolute abundance of any human protein with the high quantitative accuracy. Salirasib and siRNA targeting of HRAS inhibited cell proliferation, migration and invasion in HRAS wild type and HRAS-mutated cell lines. Proteomic analyses revealed that several metabolic pathways, including the oxidative phosphorylation pathway and glycolysis, were significantly downregulated in salirasib-treated BC cells. However, the expression levels of hexokinase 2, phosphoglycerate kinase 1, pyruvate kinase, muscle (PKM)1, PKM2 and lactate dehydrogenase A, which are downstream of RAS and target genes of hypoxia inducible factor-1α, were not notably downregulated, which may explain the high concentration of salirasib required to inhibit cell viability. These findings provide insight into the mechanisms of salirasib, and suggest the need for novel therapeutic strategies to treat cancers such as BC.


Subject(s)
Antineoplastic Agents/administration & dosage , Farnesol/analogs & derivatives , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Salicylates/administration & dosage , Up-Regulation/drug effects , Urinary Bladder Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Farnesol/administration & dosage , Farnesol/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Gene Regulatory Networks , Glycolysis/drug effects , Humans , Mice , Mutation , Oxidative Phosphorylation/drug effects , Proteomics , Proto-Oncogene Mas , Salicylates/pharmacology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...