Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Bioorg Chem ; 87: 163-168, 2019 06.
Article in English | MEDLINE | ID: mdl-30884310

ABSTRACT

Autophagy is an intracellular degradation/recycling pathway that provides nutrients and building blocks to cellular metabolism and keeps the cytoplasm clear of obsolete proteins and organelles. During recent years, dysregulated autophagy activity has been reported to be a characteristic of many different disease types, including cancer and neurodegenerative disorders. This has created a strong case for development of autophagy modulating compounds as potential treatments for these diseases. Inhibitors of autophagy have been proposed as a therapeutic intervention in, e.g., advanced cancer, and inhibiting the cysteine protease Atg4B has been put forward as a main strategy to block autophagy. We recently identified and demonstrated -both in vitro and in vivo - that compounds with a benzotropolone basic structure targeting Atg4B, can significantly slow down tumor growth and potentiate the effect of classical chemotherapy. In this study we report the synthesis and inhibition profile of new benzotropolone derivatives with additional structural modifications at 6 different positions. To obtain a solid inhibition profile, all compounds were evaluated on three levels, including two cell-based assays to confirm autophagy and intracellular Atg4B inhibition and an SDS-PAGE-based experiment to assess in vitro Atg4B affinity. Several molecules with a promising profile were identified.


Subject(s)
Autophagy-Related Proteins/antagonists & inhibitors , Autophagy/drug effects , Enzyme Inhibitors/pharmacology , Tropolone/pharmacology , Autophagy-Related Proteins/metabolism , Cysteine Endopeptidases/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Structure-Activity Relationship , Tropolone/analogs & derivatives , Tropolone/chemistry
2.
Vascul Pharmacol ; 113: 70-76, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30590134

ABSTRACT

BACKGROUND AND AIMS: Inhibition of the mechanistic target of rapamycin (mTOR) is a promising approach to halt atherogenesis in different animal models. This study evaluated whether the mTOR inhibitor everolimus can stabilize pre-existing plaques, prevent cardiovascular complications and improve survival in a mouse model of advanced atherosclerosis. METHODS: ApoE-/-Fbn1C1039G+/- mice (n = 24) were fed a Western diet (WD) for 12 weeks. Subsequently, mice were treated with everolimus (1.5 mg/kg daily) or vehicle for another 12 weeks while the WD continued. RESULTS: Despite hypercholesterolemia, everolimus treatment was associated with a reduction in circulating Ly6Chigh monocytes (15 vs. 28% of total leukocytes, p = 0.046), a depletion of plaque macrophages (2.1 vs. 4.1%, p = 0.040) and an abolishment of intraplaque neovascularization, which are all indicative of a more stable plaque phenotype. Moreover, everolimus reduced hypoxic brain damage and improved cardiac function, which led to increased survival (100 vs. 67% of animals, p = 0.038). CONCLUSIONS: Everolimus enhances features of plaque stability and counters cardiovascular complications in ApoE-/-Fbn1C1039G+/- mice, even when administered at a later stage of the disease.


Subject(s)
Atherosclerosis/drug therapy , Cardiovascular Agents/pharmacology , Carotid Artery Diseases/drug therapy , Carotid Artery, Common/drug effects , Everolimus/pharmacology , Macrophages/drug effects , Neovascularization, Pathologic , Plaque, Atherosclerotic , Animals , Antigens, Ly/metabolism , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Brain/drug effects , Brain/pathology , Brain/physiopathology , Carotid Artery Diseases/genetics , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/pathology , Carotid Artery, Common/metabolism , Carotid Artery, Common/pathology , Diet, Western , Disease Models, Animal , Disease Progression , Female , Fibrillin-1/deficiency , Fibrillin-1/genetics , Heart/drug effects , Heart/physiopathology , Hypoxia, Brain/pathology , Hypoxia, Brain/physiopathology , Hypoxia, Brain/prevention & control , Macrophages/metabolism , Macrophages/pathology , Mice, Knockout, ApoE , Monocytes/drug effects , Monocytes/metabolism , Motor Activity/drug effects , Myocardial Contraction/drug effects , Protein Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
3.
Transplantation ; 102(2S Suppl 1): S44-S46, 2018 02.
Article in English | MEDLINE | ID: mdl-28230638

ABSTRACT

Inhibitors of the mechanistic target of rapamycin (mTOR) have unique antiatherosclerotic effects, such as depletion of plaque macrophages, induction of autophagy, and activation of cholesterol efflux. However, a common side effect of their use is dyslipidemia, a well-known risk factor for atherosclerosis. Indeed, mTOR inhibitors prevent lipid storage, increase low-density lipoprotein cholesterol levels, and activate lipolysis. Although the net effect of mTOR inhibition seems favorable, the use of cholesterol lowering drugs to manage dyslipidemia remains the most recommended strategy.


Subject(s)
Atherosclerosis/prevention & control , Dyslipidemias/chemically induced , Everolimus/adverse effects , Protein Kinase Inhibitors/adverse effects , Sirolimus/adverse effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Atherosclerosis/chemically induced , Atherosclerosis/complications , Dyslipidemias/complications , Everolimus/therapeutic use , Humans , Protein Kinase Inhibitors/therapeutic use , Sirolimus/therapeutic use
4.
Biochem Pharmacol ; 138: 150-162, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28642033

ABSTRACT

Autophagy is a cell survival mechanism hijacked by advanced tumors to endure a rough microenvironment. Late autophagy inhibitors such as (hydroxy)chloroquine have been used clinically to halt tumor progression with modest success. However, given the toxic nature of these compounds and their lack of specificity, novel targets should be considered. We recently identified a benzotropolone derivative that significantly inhibited the essential autophagy protein ATG4B. Therefore, we synthesized and tested additional benzotropolone compounds to identify a promising ATG4B inhibitor that impairs autophagy both in vitro and in vivo. A compound library containing 27 molecules with a benzotropolone backbone was synthesized and screened for inhibition of recombinant ATG4B. Depending on the benzotropolone compound, inhibition of recombinant ATG4B ranged from 3 to 82%. Active compounds were evaluated in cellular assays to confirm inhibition of ATG4B and suppression of autophagy. Seven compounds inhibited processing of the autophagy protein LC3 and autophagosome formation. Compound UAMC-2526 was selected for further in vivo use because of its fair plasma stability. This compound abolished autophagy both in nutrient-deprived GFP-LC3 mice and in CD1-/- Foxn1nu mice bearing HT29 colorectal tumor xenografts. Moreover, addition of UAMC-2526 to the chemotherapy drug oxaliplatin significantly improved inhibition of tumor growth. Our data indicate that suppression of autophagy via ATG4B inhibition is a feasible strategy to augment existing chemotherapy efficacy and to halt tumor progression.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Autophagy-Related Proteins/antagonists & inhibitors , Autophagy/drug effects , Colonic Neoplasms/drug therapy , Cysteine Proteinase Inhibitors/therapeutic use , Drug Design , Tropolone/analogs & derivatives , Adenocarcinoma/pathology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/pharmacology , Drug Stability , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , HEK293 Cells , HT29 Cells , Humans , Jurkat Cells , Mice, Knockout , Mice, Transgenic , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Organoplatinum Compounds/therapeutic use , Oxaliplatin , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Tropolone/chemistry , Tropolone/pharmacology , Tropolone/therapeutic use , Xenograft Model Antitumor Assays
5.
Br J Pharmacol ; 173(23): 3359-3371, 2016 12.
Article in English | MEDLINE | ID: mdl-27638766

ABSTRACT

BACKGROUND AND PURPOSE: Everolimus is an allosteric inhibitor of the mechanistic target of rapamycin complex 1 (mTORC1) widely known for its potent autophagy stimulating properties. Because everolimus shows poor solubility and stability in aqueous solutions, long-term in vivo administration in preclinical models is challenging. The aim of the present study was to evaluate the effects of short-term and long-term everolimus administration on mTORC1 inhibition and autophagy induction in mice. EXPERIMENTAL APPROACH: We developed a vehicle in which everolimus was solubilized and stable at 37°C for at least 1 month. Using osmotic minipumps, GFP microtubule-associated protein light chain 3 transgenic mice were treated continuously either with vehicle or everolimus (1.5 mg·kg-1 per day) for 3 or 28 days. Alternatively, a regimen consisting of intermittent everolimus administration (every other day) for 56 days by oral gavage was used. Autophagy markers and mTORC1 activation status were investigated in the liver. KEY RESULTS: As expected, everolimus inhibited mTORC1 and stimulated autophagy in the liver after 3 days of treatment. However, continuous administration for 28 days resulted in hyperactivation of the Akt1-mTORC1 pathway accompanied by a remarkable decrease in autophagy markers. Everolimus given intermittently for 56 days partially rescued mTORC1 sensitivity to the drug but without inducing autophagy. The failure to induce autophagy following long-term everolimus administration was due to uncoupling of the mTORC1 substrate unc-51 like autophagy activating kinase 1. CONCLUSIONS AND IMPLICATIONS: Our data encourage the use of intermittent everolimus regimens to prevent tolerance and to extend its activity.


Subject(s)
Autophagy/drug effects , Drug Tolerance , Everolimus/pharmacology , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Animals , Drug Administration Schedule , Drug Stability , Everolimus/administration & dosage , Everolimus/chemistry , Female , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Liver/drug effects , Liver/metabolism , Mice , Mice, Transgenic , Solubility , Time Factors
6.
Atherosclerosis ; 251: 319-327, 2016 08.
Article in English | MEDLINE | ID: mdl-27450786

ABSTRACT

BACKGROUND AND AIMS: Spermidine is an endogenous biological polyamine that exhibits broad longevity-extending activities via the induction of autophagy. Because basal autophagy is atheroprotective during early atherosclerosis but dysfunctional in advanced plaques, the aim of the present study was to assess the potential beneficial effects of autophagy induction by spermidine on atherosclerotic plaque progression and composition. METHODS: Apolipoprotein E-deficient (ApoE(-/-)) mice were fed a Western-type diet for 20 weeks with or without 5 mM spermidine in the drinking water. RESULTS: (Immuno-)histochemical analysis of plaques in the aortic root, proximal ascending aorta and brachiocephalic artery showed that spermidine changed neither the size of the plaque nor its cellular composition. However, spermidine treatment significantly reduced necrotic core formation (6.6 ± 0.5% vs. 3.7 ± 0.5% in aortic root, p = 0.0008) and lipid accumulation inside the plaque (27 ± 3% vs. 17 ± 1% oil red O positivity in thoracic aorta, p = 0.017). In vitro experiments showed that macrophages, unlike vascular smooth muscle cells (VSMCs), were relatively insensitive to autophagy induction by spermidine. Along these lines, spermidine triggered cholesterol efflux in autophagy-competent VSMCs (5.7 ± 1.2% vs. 8.7 ± 0.2%, p = 0.0118), but not in autophagy-deficient Atg7(F/F)SM22α-Cre(+) VSMCs or macrophages. Analogous to the experiments in vitro, spermidine affected neither necrosis nor lipid load in plaques of Atg7(F/F)SM22α-Cre(+)ApoE(-/-) mice. CONCLUSIONS: Spermidine inhibits lipid accumulation and necrotic core formation through stimulation of cholesterol efflux, albeit without changing plaque size or cellular composition. These effects, which are driven by autophagy in VSMCs, support the general idea that autophagy induction is potentially useful to prevent vascular disease.


Subject(s)
Autophagy , Lipids/chemistry , Necrosis/pathology , Plaque, Atherosclerotic/pathology , Spermidine/therapeutic use , Animals , Atherosclerosis/drug therapy , Cholesterol/chemistry , Humans , Macrophages/drug effects , Male , Mice , Mice, Knockout, ApoE , Myocytes, Smooth Muscle/metabolism , Necrosis/drug therapy , Plaque, Atherosclerotic/drug therapy
7.
J Physiol ; 594(21): 6105-6115, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27256450

ABSTRACT

KEY POINTS: Cyclic stretch is known to alter intracellular pathways involved in vessel tone regulation. We developed a novel set-up that allows straightforward characterization of the biomechanical properties of the mouse aorta while stretched at a physiological heart rate (600 beats min-1 ). Active vessel tone was shown to have surprisingly large effects on isobaric stiffness. The effect of structural vessel wall alterations was confirmed using a genetic mouse model. This set-up will contribute to a better understanding of how active vessel wall components and mechanical stimuli such as stretch frequency and amplitude regulate aortic mechanics. ABSTRACT: Cyclic stretch is a major contributor to vascular function. However, isolated mouse aortas are frequently studied at low stretch frequency or even in isometric conditions. Pacing experiments in rodents and humans show that arterial compliance is stretch frequency dependent. The Rodent Oscillatory Tension Set-up to study Arterial Compliance is an in-house developed organ bath set-up that clamps aortic segments to imposed preloads at physiological rates up to 600 beats min-1 . The technique enables us to derive pressure-diameter loops and assess biomechanical properties of the segment. To validate the applicability of this set-up we aimed to confirm the effects of distension pressure and vascular smooth muscle tone on arterial stiffness. At physiological stretch frequency (10 Hz), the Peterson modulus (EP ; 293 (10) mmHg) for wild-type mouse aorta increased 22% upon a rise in pressure from 80-120 mmHg to 100-140 mmHg, while, at normal pressure, EP increased 80% upon maximal contraction of the vascular smooth muscle cells. We further validated the method using a mouse model with a mutation in the fibrillin-1 gene and an endothelial nitric oxide synthase knock-out model. Both models are known to have increased arterial stiffness, and this was confirmed using the set-up. To our knowledge, this is the first set-up that facilitates the study of biomechanical properties of mouse aortic segments at physiological stretch frequency and pressure. We believe that this set-up can contribute to a better understanding of how cyclic stretch frequency, amplitude and active vessel wall components influence arterial stiffening.


Subject(s)
Aorta/physiology , Muscle Contraction , Organ Culture Techniques/methods , Amplifiers, Electronic , Animals , Biomechanical Phenomena , Mice , Mice, Inbred C57BL , Myography/instrumentation , Myography/methods , Organ Culture Techniques/instrumentation
8.
Br J Clin Pharmacol ; 82(5): 1267-1279, 2016 11.
Article in English | MEDLINE | ID: mdl-26551391

ABSTRACT

Despite significant improvement in the management of atherosclerosis, this slowly progressing disease continues to affect countless patients around the world. Recently, the mechanistic target of rapamycin (mTOR) has been identified as a pre-eminent factor in the development of atherosclerosis. mTOR is a constitutively active kinase found in two different multiprotein complexes, mTORC1 and mTORC2. Pharmacological interventions with a class of macrolide immunosuppressive drugs, called rapalogs, have shown undeniable evidence of the value of mTORC1 inhibition to prevent the development of atherosclerotic plaques in several animal models. Rapalog-eluting stents have also shown extraordinary results in humans, even though the exact mechanism for this anti-atherosclerotic effect remains elusive. Unfortunately, rapalogs are known to trigger diverse undesirable effects owing to mTORC1 resistance or mTORC2 inhibition. These adverse effects include dyslipidaemia and insulin resistance, both known triggers of atherosclerosis. Several strategies, such as combination therapy with statins and metformin, have been suggested to oppose rapalog-mediated adverse effects. Statins and metformin are known to inhibit mTORC1 indirectly via 5' adenosine monophosphate-activated protein kinase (AMPK) activation and may hold the key to exploit the full potential of mTORC1 inhibition in the treatment of atherosclerosis. Intermittent regimens and dose reduction have also been proposed to improve rapalog's mTORC1 selectivity, thereby reducing mTORC2-related side effects.


Subject(s)
Atherosclerosis/drug therapy , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Drug Therapy, Combination , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Mechanistic Target of Rapamycin Complex 1/physiology , Metformin/therapeutic use , Models, Biological , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Signal Transduction/drug effects , Signal Transduction/physiology , TOR Serine-Threonine Kinases/physiology
9.
Circ Res ; 116(3): 468-79, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25634970

ABSTRACT

Autophagy is a reparative, life-sustaining process by which cytoplasmic components are sequestered in double-membrane vesicles and degraded on fusion with lysosomal compartments. Growing evidence reveals that basal autophagy is an essential in vivo process mediating proper vascular function. Moreover, autophagy is stimulated by many stress-related stimuli in the arterial wall to protect endothelial cells and smooth muscle cells against cell death and the initiation of vascular disease, in particular atherosclerosis. Basal autophagy is atheroprotective during early atherosclerosis but becomes dysfunctional in advanced atherosclerotic plaques. Little is known about autophagy in other vascular disorders, such as aneurysm formation, arterial aging, vascular stiffness, and chronic venous disease, even though autophagy is often impaired. This finding highlights the need for pharmacological interventions with compounds that stimulate the prosurvival effects of autophagy in the vasculature. A large number of animal studies and clinical trials have indicated that oral or stent-based delivery of the autophagy inducer rapamycin or derivatives thereof, collectively known as rapalogs, effectively inhibit the basic mechanisms that control growth and destabilization of atherosclerotic plaques. Other autophagy-inducing drugs, such as spermidine or add-on therapy with widely used antiatherogenic compounds, including statins and metformin, are potentially useful to prevent vascular disease with minimal adverse effects.


Subject(s)
Autophagy , Vascular Diseases/metabolism , Animals , Endothelium, Vascular/metabolism , Humans , Muscle, Smooth, Vascular/metabolism , Vascular Diseases/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...