Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 506
Filter
1.
Cell ; 184(2): 293, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33482093

ABSTRACT

The ultra-rare, pediatric premature aging disorder Hutchinson-Gilford progeria syndrome (HGPS) is caused by mutation of LMNA, encoding the nuclear architectural protein lamin A. Patients develop atherosclerosis and typically die of heart failure in their teens. FDA-approved Zokinvy prevents farnesylation of lamin A, reduces vascular stiffness, and extends survival in HGPS patients. To view this Bench to Bedside, open or download the PDF.


Subject(s)
Enzyme Inhibitors/therapeutic use , Farnesyltranstransferase/antagonists & inhibitors , Progeria/drug therapy , Progeria/enzymology , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/metabolism , Humans , Molecular Targeted Therapy
2.
Cell ; 156(3): 400-7, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24485450

ABSTRACT

Rare diseases are powerful windows into biological processes and can serve as models for the development of therapeutic strategies. The progress made on the premature aging disorder Progeria is a shining example of the impact that studies of rare diseases can have.


Subject(s)
Progeria/drug therapy , Progeria/physiopathology , Translational Research, Biomedical , Aging/genetics , Aging/pathology , Child , Farnesyltranstransferase/antagonists & inhibitors , Humans , Lamin Type A , Nuclear Proteins/metabolism , Progeria/genetics , Progeria/pathology , Protein Precursors/metabolism
3.
Development ; 147(12)2020 06 24.
Article in English | MEDLINE | ID: mdl-32467241

ABSTRACT

Tendons and ligaments are crucial components of the musculoskeletal system, yet the pathways specifying these fates remain poorly defined. Through a screen of known bioactive chemicals in zebrafish, we identified a new pathway regulating tendon cell induction. We established that statin, through inhibition of the mevalonate pathway, causes an expansion of the tendon progenitor population. Co-expression and live imaging studies indicate that the expansion does not involve an increase in cell proliferation, but rather results from re-specification of cells from the neural crest-derived sox9a+/sox10+ skeletal lineage. The effect on tendon cell expansion is specific to the geranylgeranylation branch of the mevalonate pathway and is mediated by inhibition of Rac activity. This work establishes a novel role for the mevalonate pathway and Rac activity in regulating specification of the tendon lineage.


Subject(s)
Mevalonic Acid/metabolism , Tendons/metabolism , Alkyl and Aryl Transferases/antagonists & inhibitors , Alkyl and Aryl Transferases/genetics , Alkyl and Aryl Transferases/metabolism , Animals , Animals, Genetically Modified/metabolism , Atorvastatin/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/genetics , Farnesyltranstransferase/metabolism , Morpholinos/metabolism , Neural Crest/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Tendons/cytology , Tendons/pathology , Zebrafish/metabolism , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/metabolism
4.
Crit Rev Biochem Mol Biol ; 54(1): 41-60, 2019 02.
Article in English | MEDLINE | ID: mdl-30773935

ABSTRACT

Dysregulation of isoprenoid biosynthesis is implicated in numerous biochemical disorders that play a role in the onset and/or progression of age-related diseases, such as hypercholesterolemia, osteoporosis, various cancers, and neurodegeneration. The mevalonate metabolic pathway is responsible for the biosynthesis of the two key isoprenoid metabolites, farnesyl pyrophosphate (FPP) and geranylgeranyl pyrophosphate (GGPP). Post-translational prenylation of various proteins, including the small GTP-binding proteins (GTPases), with either FPP or GGPP is vital for proper localization and activation of these proteins. Prenylated GTPases play a critical role in cell signaling, proliferation, cellular plasticity, oncogenesis, and cancer metastasis. Pre-clinical and clinical studies strongly suggest that inhibition of protein prenylation can be an effective treatment for non-skeletal cancers. In this review, we summarize the most recent drug discovery efforts focusing on blocking protein farnesylation and/or geranylgeranylation and the biochemical and structural data available in guiding the current on-going studies in drug discovery. Furthermore, we provide a summary on the biochemical association between disruption of protein prenylation, endoplasmic reticulum (ER) stress, unfolded protein response (UPR) signaling, and cancer.


Subject(s)
Biosynthetic Pathways/drug effects , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Geranyltranstransferase/antagonists & inhibitors , Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Discovery , Enzyme Inhibitors/therapeutic use , Farnesyltranstransferase/metabolism , Geranyltranstransferase/metabolism , Humans , Mevalonic Acid/metabolism , Models, Molecular , Neoplasms/metabolism , Polyisoprenyl Phosphates/antagonists & inhibitors , Polyisoprenyl Phosphates/metabolism , Protein Prenylation/drug effects , Sesquiterpenes/antagonists & inhibitors , Sesquiterpenes/metabolism
5.
Dev Biol ; 457(2): 215-225, 2020 01 15.
Article in English | MEDLINE | ID: mdl-30998907

ABSTRACT

Therapeutic approaches for cutaneous melanoma are flourishing, but despite promising results, there is an increasing number of reported primary or secondary resistance to the growing sets of drugs approved for therapy in the clinics. Combinatorial approaches may overcome resistance, as they may tackle specific weaknesses of melanoma cells, not sufficient on their own, but effective in combination with other therapies. The transgenic zebrafish line kita:ras develops melanoma with high frequency. At 3 dpf, transgenic kita:ras larvae show a hyperpigmentation phenotype as earliest evidence of abnormal melanocyte growth. Using this model, we performed a chemical screen based on automated detection of a reduction of melanocyte number caused by any of 1280 FDA or EMA approved drugs of the library. The analysis showed that 55 molecules were able to reduce by 60% or more the number of melanocytes per embryo. We further tested two compounds for each of the 5 classes, and a farnesyltransferase inhibitor (Lonafarnib), that inhibits an essential post-translational modification of HRAS and suppresses the hyperpigmentation phenotype. Combinations of Clotrimazole and Lonafarnib showed the most promising results in zebrafish embryos, allowing a dose reduction of both drugs. We performed validation of these observations in the metastatic human melanoma cell line A375M, and in normal human epithelial melanocytes (NHEM) in order to investigate the mechanism of action of Clotrimazole in blocking the proliferation of transformed melanocytes. Viability assay and analysis of energy metabolism in Clotrimazole treated cells show that this drug specifically affects melanoma cells in vitro and transformed melanocytes in vivo, having no effects on NHEM or wild type larvae. Similar effects were observed with another hit of the same class, Miconazole. Furthermore, we show that the effects of Clotrimazole are mediated by the inhibition of hexokinase activity, which is lethal to the abnormal metabolic profile of melanoma cells in vitro and in vivo. Thus, our study shows that the zebrafish can provide a phenotype-rich assay for fully automated screening approaches to identify drugs for synthetic lethal treatment in melanoma and suggest further testing of Clotrimazole in combinatorial treatments.


Subject(s)
Antineoplastic Agents/pharmacology , Clotrimazole/pharmacology , Melanoma/drug therapy , Piperidines/pharmacology , Pyridines/pharmacology , Skin Neoplasms/drug therapy , Animals , Animals, Genetically Modified , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Drug Screening Assays, Antitumor/methods , Farnesyltranstransferase/antagonists & inhibitors , Humans , Melanocytes/metabolism , Melanoma/metabolism , Miconazole/pharmacology , Skin Neoplasms/metabolism , Zebrafish , Melanoma, Cutaneous Malignant
6.
Biochem Biophys Res Commun ; 556: 93-98, 2021 06 04.
Article in English | MEDLINE | ID: mdl-33845310

ABSTRACT

Sepsis remains a leading cause of mortality in critically ill patients and is characterized by multi-organ dysfunction. Mitochondrial damage has been proposed to be involved in the pathophysiology of sepsis. In addition to metabolic impairments resulting from mitochondrial dysfunction, mitochondrial DNA (mtDNA) causes systemic inflammation as a damage-associated molecular pattern when it is released to the circulation. Metabolic derangements in skeletal muscle are a major complication of sepsis and negatively affects clinical outcomes of septic patients. However, limited knowledge is available about sepsis-induced mitochondrial damage in skeletal muscle. Here, we show that sepsis induced profound abnormalities in cristae structure, rupture of the inner and outer membranes and enlargement of the mitochondria in mouse skeletal muscle in a time-dependent manner, which was associated with increased plasma mtDNA levels. Farnesyltransferase inhibitor, FTI-277, prevented sepsis-induced morphological aberrations of the mitochondria, and blocked the increased plasma mtDNA levels along with improved survival. These results indicate that protein farnesylation plays a role in sepsis-induced damage of the mitochondria in mouse skeletal muscle. Our findings suggest that mitochondrial disintegrity in skeletal muscle may contribute to elevated circulating mtDNA levels in sepsis.


Subject(s)
DNA, Mitochondrial/blood , Farnesyltranstransferase/antagonists & inhibitors , Mitochondria/drug effects , Muscle, Skeletal/drug effects , Protective Agents/pharmacology , Protective Agents/therapeutic use , Sepsis/drug therapy , Animals , Male , Methionine/analogs & derivatives , Methionine/pharmacology , Mice , Mitochondria/pathology , Muscle, Skeletal/pathology , Sepsis/blood , Sepsis/pathology , Time Factors
7.
Pharmacol Res ; 167: 105528, 2021 05.
Article in English | MEDLINE | ID: mdl-33667685

ABSTRACT

Geranylgeranyl diphosphate synthase (GGDPS), an enzyme in the isoprenoid biosynthetic pathway (IBP), produces the isoprenoid (geranylgeranyl pyrophosphate, GGPP) used in protein geranylgeranylation reactions. Our prior studies utilizing triazole bisphosphonate-based GGDPS inhibitors (GGSIs) have revealed that these agents represent a novel strategy by which to induce cancer cell death, including multiple myeloma and pancreatic cancer. Statins inhibit the rate-limiting enzyme in the IBP and potentiate the effects of GGSIs in vitro. The in vivo effects of combination therapy with statins and GGSIs have not been determined. Here we evaluated the effects of combining VSW1198, a novel GGSI, with a statin (lovastatin or pravastatin) in CD-1 mice. Twice-weekly dosing with VSW1198 at the previously established maximally tolerated dose in combination with a statin led to hepatotoxicity, while once-weekly VSW1198-based combinations were feasible. No abnormalities in kidney, spleen, brain or skeletal muscle were observed with combination therapy. Combination therapy disrupted protein geranylgeranylation in vivo. Evaluation of hepatic isoprenoid levels revealed decreased GGPP levels in the single drug groups and undetectable GGPP levels in the combination groups. Additional studies with combinations using 50% dose-reductions of either VSW1198 or lovastatin revealed minimal hepatotoxicity with expected on-target effects of diminished GGPP levels and disruption of protein geranylgeranylation. Combination statin/GGSI therapy significantly slowed tumor growth in a myeloma xenograft model. Collectively, these studies are the first to demonstrate that combination IBP inhibitor therapy alters isoprenoid levels and disrupts protein geranylgeranylation in vivo as well as slows tumor growth in a myeloma xenograft model, thus providing the framework for future clinical exploration.


Subject(s)
Biosynthetic Pathways/drug effects , Diterpenes/administration & dosage , Drug Delivery Systems/methods , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Protein Prenylation/drug effects , Terpenes/metabolism , Triazoles/administration & dosage , Animals , Biosynthetic Pathways/physiology , Cell Line, Tumor , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Diterpenes/toxicity , Drug Evaluation, Preclinical/methods , Drug Therapy, Combination , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/toxicity , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/metabolism , Female , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Lovastatin/administration & dosage , Lovastatin/toxicity , Mice , Mice, Inbred NOD , Mice, SCID , Pravastatin/administration & dosage , Pravastatin/toxicity , Protein Prenylation/physiology , Terpenes/antagonists & inhibitors , Triazoles/toxicity , Xenograft Model Antitumor Assays/methods
8.
Bioorg Med Chem Lett ; 41: 127978, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33766764

ABSTRACT

A series of novel thiazole-containing amides were synthesized. A structure-activity relationship study of these compounds led to the identification of potent and selective PfFPPS/GGPPS inhibitors with good in vitro ADME profiles. The most promising candidate molecules were progressed to mouse in vivo PK studies and demonstrated adequate free drug exposure to warrant further investigation.


Subject(s)
Antimalarials/pharmacology , Diphosphonates/pharmacology , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Geranyltranstransferase/antagonists & inhibitors , Plasmodium falciparum/drug effects , Antimalarials/chemical synthesis , Antimalarials/chemistry , Diphosphonates/chemical synthesis , Diphosphonates/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Farnesyltranstransferase/metabolism , Geranyltranstransferase/metabolism , Molecular Structure , Parasitic Sensitivity Tests , Plasmodium falciparum/enzymology , Structure-Activity Relationship
9.
Org Biomol Chem ; 19(10): 2213-2223, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33349821

ABSTRACT

Photoactivatable protecting groups (PPGs) are useful for a broad range of applications ranging from biology to materials science. In chemical biology, induction of biological processes via photoactivation is a powerful strategy for achieving spatiotemporal control. The importance of cysteine, glutathione, and other bioactive thiols in regulating protein structure/activity and cell redox homeostasis makes modulation of thiol activity particularly useful. One major objective for enhancing the utility of photoactivatable protecting groups (PPGs) in living systems is creating PPGs with longer wavelength absorption maxima and efficient two-photon (TP) absorption. Toward these objectives, we developed a carboxyl- and dimethylamine-functionalized nitrodibenzofuran PPG scaffold (cDMA-NDBF) for thiol photoactivation, which has a bathochromic shift in the one-photon absorption maximum from λmax = 315 nm with the unfunctionalized NDBF scaffold to λmax = 445 nm. While cDMA-NDBF-protected thiols are stable in the presence of UV irradiation, they undergo efficient broad-spectrum TP photolysis at wavelengths as long as 900 nm. To demonstrate the wavelength orthogonality of cDMA-NDBF and NDBF photolysis in a biological setting, caged farnesyltransferase enzyme inhibitors (FTI) were prepared and selectively photoactivated in live cells using 850-900 nm TP light for cDMA-NDBF-FTI and 300 nm UV light for NDBF-FTI. These experiments represent the first demonstration of thiol photoactivation at wavelengths above 800 nm. Consequently, cDMA-NDBF-caged thiols should have broad applicability in a wide range of experiments in chemical biology and materials science.


Subject(s)
Benzofurans/chemistry , Enzyme Inhibitors/pharmacology , Sulfhydryl Compounds/pharmacology , Animals , Benzofurans/chemical synthesis , Benzofurans/radiation effects , Dogs , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/radiation effects , Farnesyltranstransferase/antagonists & inhibitors , Infrared Rays , Madin Darby Canine Kidney Cells , Photolysis/radiation effects , Photons , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/radiation effects
10.
Bioorg Med Chem ; 44: 116307, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34298413

ABSTRACT

Agents that inhibit the enzyme geranylgeranyl diphosphate synthase (GGDPS) have anti-cancer activity and our prior studies have investigated the structure-function relationship for a family of isoprenoid triazole bisphosphonates as GGDPS inhibitors. To further explore this structure-function relationship, a series of novel α-modified triazole phosphonates was prepared and evaluated for activity as GGDPS inhibitors in enzyme and cell-based assays. These studies revealed flexibility at the α position of the bisphosphonate derivatives with respect to being able to accommodate a variety of substituents without significantly affecting potency compared to the parent unsubstituted inhibitor. However, the monophosphonate derivatives lacked activity. These studies further our understanding of the structure-function relationship of the triazole-based GGDPS inhibitors and lay the foundation for future studies evaluating the impact of α-modifications on in vivo activity.


Subject(s)
Diphosphonates/pharmacology , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Triazoles/pharmacology , Diphosphonates/chemical synthesis , Diphosphonates/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Farnesyltranstransferase/metabolism , Humans , Molecular Structure , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/chemistry
11.
Bioorg Med Chem ; 51: 116513, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34798379

ABSTRACT

A series of new quinazolinedione derivatives have been readily synthesized and evaluated for their in vitro antiplasmodial growth inhibition activity. Most of the compounds inhibited P. falciparum FcB1 strain in the low to medium micromolar concentration. The 2-ethoxy 8ag', 2-trifluoromethoxy 8ai' and 4-fluoro-2-methoxy 8ak' showed the best inhibitory activity with EC50 values around 5 µM and were non-toxic to the primary human fibroblast cell line AB943. However, these compounds were less potent than the original hit MMV665916, which showed remarkable growth inhibition with EC50 value of 0.4 µM and presented the highest selectivity index (SI > 250). In addition, a novel approach for determining the docking poses of these quinazolinedione derivatives with their potential protein target, the P. falciparum farnesyltransferase PfFT, was investigated.


Subject(s)
Antimalarials/pharmacology , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Plasmodium falciparum/drug effects , Antimalarials/chemical synthesis , Antimalarials/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Farnesyltranstransferase/metabolism , Models, Molecular , Molecular Structure , Parasitic Sensitivity Tests , Plasmodium falciparum/enzymology , Structure-Activity Relationship
12.
Semin Cancer Biol ; 56: 128-134, 2019 06.
Article in English | MEDLINE | ID: mdl-29100957

ABSTRACT

Ras proteins have been reported to play key role in oncologic diseases. Ras proteins are associated with cellular membranes for its carcinogenic activities through post-translational modifications, including farnesylation. Farnesyltransferase is responsible for a type of Ras membrane targeting, which leads to cancer origin and progression. Inhibitors of farnesyltransferase have been developed as novel anticancer agents. In this review, the role of farnesyltransferase in cancer progression and development has been discussed. Further, the current status of development of farnesyltransferase inhibitors for cancer prevention and treatment has also been reviewed.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/metabolism , Neoplasms/metabolism , Animals , Antineoplastic Agents/therapeutic use , Clinical Trials as Topic , Enzyme Inhibitors/therapeutic use , Gene Expression Regulation, Neoplastic , Genes, ras , Humans , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/etiology , Neoplasms/pathology , Protein Binding , Signal Transduction , Treatment Outcome
13.
Br J Cancer ; 123(12): 1749-1756, 2020 12.
Article in English | MEDLINE | ID: mdl-32968206

ABSTRACT

BACKGROUND: Multi-targeted tyrosine kinase inhibitors (TKIs) are the standard of care for patients with advanced clear cell renal cell carcinoma (ccRCC). However, a significant number of ccRCC patients are primarily refractory to targeted therapeutics, showing neither disease stabilisation nor clinical benefits. METHODS: We used CRISPR/Cas9-based high-throughput loss of function (LOF) screening to identify cellular factors involved in the resistance to sunitinib. Next, we validated druggable molecular factors that are synthetically lethal with sunitinib treatment using cell and animal models of ccRCC. RESULTS: Our screening identified farnesyltransferase among the top hits contributing to sunitinib resistance in ccRCC. Combined treatment with farnesyltransferase inhibitor lonafarnib potently augmented the anti-tumour efficacy of sunitinib both in vitro and in vivo. CONCLUSION: CRISPR/Cas9 LOF screening presents a promising approach to identify and target cellular factors involved in the resistance to anti-cancer therapeutics.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/drug therapy , Drug Resistance, Neoplasm/genetics , Farnesyltranstransferase/antagonists & inhibitors , Kidney Neoplasms/drug therapy , Piperidines/pharmacology , Pyridines/pharmacology , Sunitinib/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Apoptosis , CRISPR-Cas Systems , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , DNA Fragmentation , Drug Interactions , Drug Therapy, Combination , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Lysosomes , Male , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Molecular Targeted Therapy , Neoplasm Transplantation , Progression-Free Survival , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering , Random Allocation , Sunitinib/pharmacokinetics
14.
Bioorg Med Chem Lett ; 30(11): 127149, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32247731

ABSTRACT

A broad range of chalcones and derivatives were easily and rapidly synthesized, following Claisen-Schmidt condensation of (hetero)aryl ketones and (hetero)aryl aldehydes using a ultrasound probe. A comparison was made with classical magnetic stirring experiments, and an optimization study was realized, showing lithium hydroxide to be the best basic catalyst of the studied condensations. By-products of the reactions (ß-hydroxy-ketone, diketones, and cyclohexanols) were also isolated. All compounds were evaluated in vitro for their ability to inhibit human farnesyltransferase, a protein implicated in cancer and rare diseases and on the NCI-60 cancer cell lines panel. Molecules showed inhibitory activity on the target protein and cytostatic effect on different cell lines with particular activity against MCF7, breast cancer cells.


Subject(s)
Antineoplastic Agents/chemical synthesis , Chalcones/chemistry , Enzyme Inhibitors/chemical synthesis , Farnesyltranstransferase/antagonists & inhibitors , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Binding Sites , Catalytic Domain , Cell Line, Tumor , Cell Survival/drug effects , Chalcones/metabolism , Chalcones/pharmacology , Drug Screening Assays, Antitumor , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/metabolism , Humans , Molecular Docking Simulation , Sonication , Structure-Activity Relationship
15.
Bioorg Med Chem ; 28(16): 115604, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32690260

ABSTRACT

Geranylgeranyl diphosphate synthase (GGDPS) inhibitors are of potential therapeutic interest as a consequence of their activity against the bone marrow cancer multiple myeloma. A series of bisphosphonates linked to an isoprenoid tail through an amide linkage has been prepared and tested for the ability to inhibit GGDPS in enzyme and cell-based assays. The amides were designed as analogues to triazole-based GGDPS inhibitors. Several of the new compounds show GGDPS inhibitory activity in both enzyme and cell assays, with potency dependent on chain length and olefin stereochemistry.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Triazoles/chemistry , Triazoles/pharmacology , Amides/chemistry , Amides/pharmacology , Cell Line , Diphosphonates/chemistry , Diphosphonates/pharmacology , Farnesyltranstransferase/metabolism , Humans , Models, Molecular , Structure-Activity Relationship , Terpenes/chemistry , Terpenes/pharmacology
16.
J Immunol ; 200(11): 3840-3856, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29703864

ABSTRACT

Ras, a small GTPase protein, is thought to mediate Th2-dependent eosinophilic inflammation in asthma. Ras requires cell membrane association for its biological activity, and this requires the posttranslational modification of Ras with an isoprenyl group by farnesyltransferase (FTase) or geranylgeranyltransferase (GGTase). We hypothesized that inhibition of FTase using FTase inhibitor (FTI)-277 would attenuate allergic asthma by depleting membrane-associated Ras. We used the OVA mouse model of allergic inflammation and human airway epithelial (HBE1) cells to determine the role of FTase in inflammatory cell recruitment. BALB/c mice were first sensitized then exposed to 1% OVA aerosol or filtered air, and half were injected daily with FTI-277 (20 mg/kg per day). Treatment of mice with FTI-277 had no significant effect on lung membrane-anchored Ras, Ras protein levels, or Ras GTPase activity. In OVA-exposed mice, FTI-277 treatment increased eosinophilic inflammation, goblet cell hyperplasia, and airway hyperreactivity. Human bronchial epithelial (HBE1) cells were pretreated with 5, 10, or 20 µM FTI-277 prior to and during 12 h IL-13 (20 ng/ml) stimulation. In HBE1 cells, FTase inhibition with FTI-277 had no significant effect on IL-13-induced STAT6 phosphorylation, eotaxin-3 peptide secretion, or Ras translocation. However, addition of exogenous FPP unexpectedly augmented IL-13-induced STAT6 phosphorylation and eotaxin-3 secretion from HBE1 cells without affecting Ras translocation. Pharmacological inhibition of FTase exacerbates allergic asthma, suggesting a protective role for FTase or possibly Ras farnesylation. FPP synergistically augments epithelial eotaxin-3 secretion, indicating a novel Ras-independent farnesylation mechanism or direct FPP effect that promotes epithelial eotaxin-3 production in allergic asthma.


Subject(s)
Asthma/drug therapy , Bronchial Hyperreactivity/drug therapy , Eosinophils/drug effects , Farnesyltranstransferase/antagonists & inhibitors , Inflammation/drug therapy , Polyisoprenyl Phosphates/metabolism , Sesquiterpenes/metabolism , ras Proteins/metabolism , Animals , Asthma/metabolism , Bronchi/drug effects , Bronchi/metabolism , Bronchial Hyperreactivity/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Eosinophils/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Farnesyltranstransferase/metabolism , Humans , Inflammation/metabolism , Lung/drug effects , Lung/metabolism , Male , Methionine/analogs & derivatives , Methionine/pharmacology , Mice , Mice, Inbred BALB C , Ovalbumin/pharmacology , Signal Transduction/drug effects
17.
Bioorg Chem ; 103: 104184, 2020 10.
Article in English | MEDLINE | ID: mdl-32891861

ABSTRACT

In the incessant search for innovative cancer control strategies, this study was devoted to the design, synthesis and pharmacological evaluation of dual inhibitors of farnesyltransferase and tubulin polymerization (FTI/MTIs). A series of indolizine-phenothiazine hybrids 16 (amides) and 17 (ketones) has been obtained in a 4-step procedure. The combination of the two heterocycles provided potent tubulin polymerization inhibitors with similar efficiency as the reference phenstatin and (-)-desoxypodophyllotoxin. Ketones 17 were also able to inhibit human farnesyltransferase (FTase) in vitro. Interestingly, three molecules 17c, 17d and 17f were very effective against both considered biological targets. Next, nine indolizine-phenothiazine hybrids 16c, 16f, 17a-f and 22b were evaluated for their cell growth inhibition potential on the NCI-60 cancer cell lines panel. Ketones 17a-f were the most active and displayed promising cellular activities. Not only they arrested the cell growth of almost all tested cancer cells, but they displayed cytotoxicity potential with GI50 values in the low nanomolar range. The most sensitive cell lines upon treatment with indolizine-phenothiazine hybrids were NCI-H522 (lung cancer), COLO-205 and HT29 (colon cancer), SF-539 (human glioblastoma), OVCAR-3 (ovarian cancer), A498 (renal cancer) and especially MDA-MB-435 (melanoma). Demonstrating the preclinical effectiveness of these dual inhibitors can be crucial. A single dual molecule could induce a synergy of antitumor activity, while increasing the effectiveness and reducing the toxicity of the classical combo treatments currently used in chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Indolizines/pharmacology , Phenothiazines/pharmacology , Tubulin Modulators/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Farnesyltranstransferase/chemistry , Farnesyltranstransferase/metabolism , Humans , Indolizines/chemical synthesis , Indolizines/metabolism , Molecular Docking Simulation , Molecular Structure , Phenothiazines/chemical synthesis , Phenothiazines/metabolism , Protein Binding , Structure-Activity Relationship , Tubulin/chemistry , Tubulin/metabolism , Tubulin Modulators/chemical synthesis , Tubulin Modulators/metabolism
18.
J Enzyme Inhib Med Chem ; 35(1): 1027-1044, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32308053

ABSTRACT

As one of the world's five terminally ills, tumours can cause important genetic dysfunction. However, some current medicines for tumours usually have strong toxic side effects and are prone to drug resistance. Studies have found that farnesyltransferase inhibitors (FTIs) extracted from natural materials have a good inhibiting ability on tumours with fewer side effects. This article describes several FTIs extracted from natural materials and clarifies the current research progress, which provides a new choice for the treatment of tumours.


Subject(s)
Antineoplastic Agents/pharmacology , Biological Products/pharmacology , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Biological Products/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Farnesyltranstransferase/metabolism , Humans , Molecular Structure , Neoplasms/metabolism , Structure-Activity Relationship
19.
J Pharmacol Exp Ther ; 371(2): 327-338, 2019 11.
Article in English | MEDLINE | ID: mdl-31420526

ABSTRACT

The enzyme geranylgeranyl diphosphate synthase (GGDPS) synthesizes the 20-carbon isoprenoid geranylgeranyl pyrophosphate, which is used in geranylgeranylation reactions. We have demonstrated that GGDPS inhibitors in multiple myeloma (MM) cells disrupt Rab geranylgeranylation, leading to inhibition of monoclonal protein trafficking, induction of the unfolded protein response pathway (UPR), and apoptosis. We have previously reported preclinical studies with the GGDPS inhibitor VSW1198, which is a mixture of homogeranyl/homoneryl triazole bisphosphonates. Additional structure-function efforts have led to development of the α-methylated derivatives RAM2093 (homogeranyl) and RAM2061 (homoneryl). As little is known regarding the impact of olefin stereochemistry on drug properties in vivo, we pursued additional preclinical evaluation of RAM2093 and RAM2061. In MM cell lines, both isomers induce activation of UPR/apoptotic markers in a concentration-dependent manner and with similar potency. Single-dose testing in CD-1 mice identified a maximum tolerated i.v. dose of 0.5 mg/kg for RAM2061 and 0.3 mg/kg for RAM2093. Liver toxicity was the primary barrier to dose escalation for both compounds. Disruption of geranylgeranylation in vivo was confirmed after multidose administration of either compound. Pharmacokinetic studies revealed plasma terminal half-lives of 29.2 ± 6 (RAM2061) and 22.1 ± 4 hours (RAM2093). Relative to RAM2061, RAM2093 levels were significantly higher in liver tissue but not in other tissues. Using MM.1S flank xenografts, we observed a significant reduction in tumor growth in mice treated with RAM2061 relative to controls. Collectively, these studies reveal olefin stereochemistry-dependent effects on GGDPS inhibitor biodistribution and confirm the in vivo efficacy of this novel therapeutic approach. SIGNIFICANCE STATEMENT: These studies reveal olefin stereochemistry-dependent effects on the in vivo properties of two novel triazole bisphosphonate inhibitors of geranylgeranyl diphosphate synthase and demonstrate the therapeutic potential of this class of inhibitors for the treatment of multiple myeloma.


Subject(s)
Alkenes/pharmacology , Diphosphonates/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Terpenes/pharmacology , Tissue Distribution/drug effects , Triazoles/pharmacology , Alkenes/chemistry , Alkenes/metabolism , Animals , Diphosphonates/chemistry , Diphosphonates/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Farnesyltranstransferase/metabolism , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Stereoisomerism , Terpenes/chemistry , Terpenes/metabolism , Tissue Distribution/physiology , Triazoles/chemistry , Triazoles/metabolism , Xenograft Model Antitumor Assays/methods
20.
Mol Carcinog ; 58(11): 2052-2064, 2019 11.
Article in English | MEDLINE | ID: mdl-31397499

ABSTRACT

Recent studies have indicated that using statins to inhibit the mevalonate pathway induces mutant p53 degradation by impairing the interaction of mutant p53 with DnaJ subfamily A member 1 (DNAJA1). However, the role of the C-terminus of DNAJA1 with a CAAX box for farnesylation in the binding, folding, and translocation of client proteins such as mutant p53 is not known. In the present study, we used a genetically engineered mouse model of pancreatic carcinoma and showed that atorvastatin significantly increased animal survival and inhibited pancreatic carcinogenesis. There was a dramatic decrease in mutant p53 protein accumulation in the pancreatic acini, pancreas intraepithelial neoplasia lesions, and adenocarcinoma. Supplementation with farnesyl pyrophosphate, a substrate for protein farnesylation, rescued atorvastatin-induced mutant p53 degradation in pancreatic cancer cells. Tipifarnib, a farnesyltransferase inhibitor, mirrored atorvastatin's effects on mutant p53, degraded mutant p53 in a dose-dependent manner, and converted farnesylated DNAJA1 into unfarnesylated DNAJA1. Farnesyltransferase gene knockdown also significantly promoted mutant p53 degradation. Coimmunoprecipitation either by an anti-DNAJA1 or p53 antibody confirmed the direct interaction of mutant p53 and DNAJA1 and higher doses of atorvastatin treatments converted more farnesylated DNAJA1 into unfarnesylated DNAJA1 with much less mutant p53 pulled down by DNAJA1. Strikingly, C394S mutant DNAJA1, in which the cysteine of the CAAX box was mutated to serine, was no longer able to be farnesylated and lost the ability to maintain mutant p53 stabilization. Our results show that farnesylated DNAJA1 is a crucial chaperone in maintaining mutant p53 stabilization and targeting farnesylated DNAJA1 by atorvastatin will be critical for inhibiting p53 mutant cancer.


Subject(s)
Atorvastatin/pharmacology , HSP40 Heat-Shock Proteins/genetics , Pancreatic Neoplasms/drug therapy , Tumor Suppressor Protein p53/genetics , Animals , Carcinogenesis/drug effects , Cell Line, Tumor , Disease Models, Animal , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Molecular Chaperones/genetics , Mutant Proteins/genetics , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Prenylation/drug effects , Proto-Oncogene Proteins p21(ras)/genetics , Quinolones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL