Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
Add more filters










Publication year range
1.
J Med Chem ; 64(16): 12322-12358, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34378914

ABSTRACT

Rhodesain is a major cysteine protease of Trypanosoma brucei rhodesiense, a pathogen causing Human African Trypanosomiasis, and a validated drug target. Recently, we reported the development of α-halovinylsulfones as a new class of covalent reversible cysteine protease inhibitors. Here, α-fluorovinylsulfones/-sulfonates were optimized for rhodesain based on molecular modeling approaches. 2d, the most potent and selective inhibitor in the series, shows a single-digit nanomolar affinity and high selectivity toward mammalian cathepsins B and L. Enzymatic dilution assays and MS experiments indicate that 2d is a slow-tight binder (Ki = 3 nM). Furthermore, the nonfluorinated 2d-(H) shows favorable metabolism and biodistribution by accumulation in mice brain tissue after intraperitoneal and oral administration. The highest antitrypanosomal activity was observed for inhibitors with an N-terminal 2,3-dihydrobenzo[b][1,4]dioxine group and a 4-Me-Phe residue in P2 (2e/4e) with nanomolar EC50 values (0.14/0.80 µM). The different mechanisms of reversible and irreversible inhibitors were explained using QM/MM calculations and MD simulations.


Subject(s)
Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Sulfones/pharmacology , Sulfonic Acids/pharmacology , Trypanocidal Agents/pharmacology , Vinyl Compounds/pharmacology , Animals , Cysteine Endopeptidases/chemistry , Cysteine Proteinase Inhibitors/chemical synthesis , Cysteine Proteinase Inhibitors/metabolism , Cysteine Proteinase Inhibitors/toxicity , Enzyme Assays , Female , HeLa Cells , Humans , Kinetics , Male , Mice , Molecular Docking Simulation , Molecular Structure , Parasitic Sensitivity Tests , Protein Binding , Structure-Activity Relationship , Sulfones/chemical synthesis , Sulfones/metabolism , Sulfones/toxicity , Sulfonic Acids/chemical synthesis , Sulfonic Acids/metabolism , Sulfonic Acids/toxicity , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/metabolism , Trypanocidal Agents/toxicity , Trypanosoma brucei brucei/drug effects , Vinyl Compounds/chemical synthesis , Vinyl Compounds/metabolism , Vinyl Compounds/toxicity
2.
Exp Parasitol ; 219: 108032, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33137308

ABSTRACT

Chagas disease affects several countries around the world with health and sanitation problems. Cysteine proteases are essential for the virulence and replication of the Trypanosoma cruzi, being modulated by dipeptidyl nitriles and derivatives. Here, four dipeptidyl nitrile derivatives were assayed in three T. cruzi morphologies and two strains (Tulahuen and Y) using a set of assays: (i) analysis of the inhibitory activity against cysteine proteases; (ii) determination of the cytotoxic activity and selectivity index; (iii) verification of the inhibition of the trypomastigote invasion in the host cell. These compounds could inhibit the activity of cysteine proteases using the selective substrate Z-FR-MCA for the trypomastigote lysate and extracellular amastigotes. Interestingly, these compounds did not present relevant enzymatic inhibition for the epimastigote lysate. Most of the substances were also cytotoxic and selective against the trypomastigotes and intracellular amastigotes. The best compound of the series (Neq0662) could reduce the enzymatic activity of the cysteine proteases for the trypomastigotes and amastigotes. It was equipotent to the benznidazole drug in the cytotoxic studies using these two parasite forms. Neq0662 was also selective for the parasite, and it inhibited the invasion of the mammalian host cell in all conditions tested at 10 µM. The stereochemistry of the trifluoromethyl group was an important factor for the bioactivity when the two diastereomers (Neq0662 and Neq0663) were compared. All-in-all, these results indicate that these compounds could move further in the drug development stage because of its promising bioactive profile.


Subject(s)
Cysteine Proteinase Inhibitors/pharmacology , Nitriles/pharmacology , Trypanosoma cruzi/drug effects , Analysis of Variance , Animals , Antiparasitic Agents/chemistry , Antiparasitic Agents/pharmacology , Area Under Curve , Cell Line , Cell Survival , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/toxicity , Haplorhini , Kidney/cytology , Nitriles/chemistry , Proteolysis , Stereoisomerism , Tetrazolium Salts , Thiazoles , Trypanosoma cruzi/enzymology , Trypanosoma cruzi/growth & development , Trypanosoma cruzi/metabolism
3.
J Am Chem Soc ; 142(25): 10899-10904, 2020 06 24.
Article in English | MEDLINE | ID: mdl-32479075

ABSTRACT

Optimization of small-molecule probes or drugs is a synthetically lengthy, challenging, and resource-intensive process. Lack of automation and reliance on skilled medicinal chemists is cumbersome in both academic and industrial settings. Here, we demonstrate a high-throughput hit-to-lead process based on the biocompatible sulfur(VI) fluoride exchange (SuFEx) click chemistry. A high-throughput screening hit benzyl (cyanomethyl)carbamate (Ki = 8 µM) against a bacterial cysteine protease SpeB was modified with a SuFExable iminosulfur oxydifluoride [RN═S(O)F2] motif, rapidly diversified into 460 analogs in overnight reactions, and the products were directly screened to yield drug-like inhibitors with 480-fold higher potency (Ki = 18 nM). We showed that the improved molecule is active in a bacteria-host coculture. Since this SuFEx linkage reaction succeeds on picomole scale for direct screening, we anticipate our methodology can accelerate the development of robust biological probes and drug candidates.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Exotoxins/antagonists & inhibitors , Sulfur Compounds/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Catalytic Domain , Click Chemistry , Crystallography, X-Ray , Cysteine Proteinase Inhibitors/metabolism , Cysteine Proteinase Inhibitors/toxicity , Drug Discovery , Exotoxins/chemistry , Exotoxins/metabolism , High-Throughput Screening Assays , Humans , Jurkat Cells , Microsomes, Liver/metabolism , Proof of Concept Study , Protein Binding
4.
J Med Chem ; 63(7): 3763-3783, 2020 04 09.
Article in English | MEDLINE | ID: mdl-32189500

ABSTRACT

The immunoproteasome (iP), an inducible proteasome variant harboring three immunosubunits, low molecular mass polypeptide-2 (LMP2), multicatalytic endopeptidase complex subunit-1, and low molecular mass polypeptide-7 (LMP7), is involved in multiple facets of inflammatory responses. We recently reported that YU102, a dual inhibitor of the iP subunit LMP2 and the constitutive proteasome catalytic subunit ß1, ameliorates cognitive impairments in mouse models of Alzheimer's disease (AD) independently of amyloid deposits. To investigate whether inhibition of LMP2 is sufficient to improve the cognitive functions of AD mice, here we prepared 37 YU102 analogues and identified a potent LMP2 inhibitor DB-310 (28) (IC50: 80.6 nM) with improved selectivity and permeability in cells overexpressing ABCB1 transporters. We show that DB-310 induces suppression of IL-1α production in microglia cells and improves cognitive functions in the Tg2576 transgenic mouse model of AD. This study supports that inhibition of LMP2 is a promising therapeutic strategy for treatment of AD.


Subject(s)
Alzheimer Disease/drug therapy , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/therapeutic use , Nootropic Agents/therapeutic use , Oligopeptides/therapeutic use , Animals , Cell Line, Transformed , Cysteine Proteinase Inhibitors/chemical synthesis , Cysteine Proteinase Inhibitors/toxicity , Epithelial-Mesenchymal Transition/drug effects , Humans , Interleukin-1alpha/metabolism , Mice, Transgenic , Microglia/drug effects , Molecular Structure , Nootropic Agents/chemical synthesis , Nootropic Agents/toxicity , Oligopeptides/chemical synthesis , Oligopeptides/toxicity , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/therapeutic use , Small Molecule Libraries/toxicity , Structure-Activity Relationship
5.
Am J Physiol Cell Physiol ; 316(5): C641-C648, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30789753

ABSTRACT

Insensitivity to the antiobesity hormone, leptin, has been suggested to be involved in the pathogenesis of obesity. However, the pathological mechanisms underlying the development of leptin resistance are not well-understood. This study aimed to examine the pathological mechanisms of leptin resistance in obesity. In the present study, we found that 4-hydroxy-2-nonenal (4-HNE), an aldehyde, may be involved in the development of leptin resistance. The SH-SY5Y-Ob-Rb human neuroblastoma cell line, transfected to express the Ob-Rb leptin receptor stably, was treated with 4-HNE, and leptin-induced signal transduction was analyzed. We found that 4-HNE dose- and time-dependently inhibited leptin-induced signal transducer and activator of transcription 3 (STAT3) phosphorylation, a major antiobesity signal of leptin. On the other hand, 4-HNE did not affect tyrosine phosphorylation of broad cellular proteins, suggesting that the inhibitory effect may be selective to leptin signaling. Mechanistically, 4-HNE induced the eukaryotic initiation factor 2α-CCAAT/enhancer-binding protein homologous protein arm of endoplasmic reticulum stress signaling, which may be involved in the pathogenesis of leptin resistance. Overall, these results suggest that 4-HNE may partly affect endoplasmic reticulum stress-induced unfolded protein response signaling and may be involved in the pathogenesis of leptin resistance.


Subject(s)
Aldehydes/toxicity , Cysteine Proteinase Inhibitors/toxicity , Endoplasmic Reticulum Stress/physiology , Leptin/metabolism , Obesity/metabolism , Receptors, Leptin/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Endoplasmic Reticulum Stress/drug effects , Humans , Leptin/antagonists & inhibitors
6.
Neurotox Res ; 34(3): 706-716, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30129004

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder of the central nervous system (CNS) caused by a progressive loss of nigrostriatal dopaminergic neurons. Dysfunction of the ubiquitin-proteasome system (UPS) plays an important role in the pathogenesis of PD. Intranigral administration of the UPS inhibitor lactacystin is used to obtain a valuable animal model for investigating putative neuroprotective treatments for PD. 1-Methyl-1,2,3,4-tetrahydroisoquinoline (1MeTIQ) is an endogenous amine that displays neuroprotective properties. This compound acts as a reversible monoamine oxidase (MAO) inhibitor and a natural free radical scavenger. In the present experiment, we investigated the effect of acute and chronic treatment with 1MeTIQ on locomotor activity and the release of dopamine as well as its metabolites in the striatum of unilaterally lactacystin-lesioned and sham-operated rats using in vivo microdialysis. Additionally, changes in the level of tyrosine hydroxylase (TH) in the substantia nigra were measured. Unilateral lactacystin injection into the substantia nigra caused significant impairment of dopamine release (approx. 45%) and a marked decline in the TH level. These effects were completely antagonized by multiple treatments with 1MeTIQ. The results obtained from the in vivo microdialysis study as well as from the ex vivo experiments suggest that multiple administration of 1MeTIQ protects dopaminergic neurons against the lactacystin-induced decline in TH concentration in the substantia nigra and prevents disturbances of dopamine release in the striatum. We have demonstrated that 1MeTIQ is capable of maintaining the physiological functions of the striatal dopamine neurons damaged by unilateral lactacystin lesion.


Subject(s)
Brain Injuries/metabolism , Brain Injuries/prevention & control , Brain/drug effects , Dopamine/metabolism , Neuroprotective Agents/therapeutic use , Tetrahydroisoquinolines/therapeutic use , 3,4-Dihydroxyphenylacetic Acid/metabolism , Acetylcysteine/analogs & derivatives , Acetylcysteine/toxicity , Analysis of Variance , Animals , Brain/metabolism , Brain Injuries/chemically induced , Brain Injuries/pathology , Cysteine Proteinase Inhibitors/toxicity , Exploratory Behavior/drug effects , Functional Laterality/drug effects , Locomotion/drug effects , Male , Microdialysis , Rats , Rats, Wistar
7.
Chem Biol Drug Des ; 92(3): 1585-1596, 2018 09.
Article in English | MEDLINE | ID: mdl-29729080

ABSTRACT

Chemotherapy is currently the only effective approach to treat all forms of leishmaniasis. However, its effectiveness is severely limited due to high toxicity, long treatment length, drug resistance, or inadequate mode of administration. As a consequence, there is a need to identify new molecular scaffolds and targets as potential therapeutics for the treatment of this disease. We report a small series of 1,2-substituted-1H-benzo[d]imidazole derivatives (9a-d) showing affinity in the submicromolar range (Ki  = 0.15-0.69 µM) toward Leishmania mexicanaCPB2.8ΔCTE, one of the more promising targets for antileishmanial drug design. The compounds confirmed activity in vitro against intracellular amastigotes of Leishmania infantum with the best result being obtained with derivative 9d (IC50  = 6.8 µM), although with some degree of cytotoxicity (CC50  = 8.0 µM on PMM and CC50  = 32.0 µM on MCR-5). In silico molecular docking studies and ADME-Tox properties prediction were performed to validate the hypothesis of the interaction with the intended target and to assess the drug-likeness of these derivatives.


Subject(s)
Benzimidazoles/chemistry , Cysteine Proteases/metabolism , Cysteine Proteinase Inhibitors/chemistry , Leishmania mexicana/enzymology , Protozoan Proteins/antagonists & inhibitors , Antiprotozoal Agents/chemical synthesis , Antiprotozoal Agents/metabolism , Antiprotozoal Agents/therapeutic use , Antiprotozoal Agents/toxicity , Benzimidazoles/metabolism , Benzimidazoles/therapeutic use , Benzimidazoles/toxicity , Binding Sites , Cell Line , Cell Survival/drug effects , Cysteine Proteases/chemistry , Cysteine Proteinase Inhibitors/metabolism , Cysteine Proteinase Inhibitors/therapeutic use , Cysteine Proteinase Inhibitors/toxicity , Drug Evaluation, Preclinical , Enzyme Assays , Humans , Hydrogen Bonding , Inhibitory Concentration 50 , Leishmaniasis/drug therapy , Molecular Docking Simulation , Protein Structure, Tertiary , Protozoan Proteins/metabolism
8.
J Med Chem ; 61(8): 3370-3388, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29590751

ABSTRACT

Macrocyclic inhibitors of rhodesain (RD), a parasitic cysteine protease and drug target for the treatment of human African trypanosomiasis, have shown low metabolic stability at the macrocyclic ether bridge. A series of acyclic dipeptidyl nitriles was developed using structure-based design (PDB ID: 6EX8 ). The selectivity against the closely related cysteine protease human cathepsin L (hCatL) was substantially improved, up to 507-fold. In the S2 pocket, 3,4-dichlorophenylalanine residues provided high trypanocidal activities. In the S3 pocket, aromatic residues provided enhanced selectivity against hCatL. RD inhibition ( Ki values) and in vitro cell-growth of Trypanosoma brucei rhodesiense (IC50 values) were measured in the nanomolar range. Triazole-based ligands, obtained by a safe, gram-scale flow production of ethyl 1 H-1,2,3-triazole-4-carboxylate, showed excellent metabolic stability in human liver microsomes and in vivo half-lives of up to 1.53 h in mice. When orally administered to infected mice, parasitaemia was reduced but without complete removal of the parasites.


Subject(s)
Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/therapeutic use , Dipeptides/therapeutic use , Nitriles/therapeutic use , Triazoles/therapeutic use , Trypanocidal Agents/therapeutic use , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Binding Sites , Cell Line , Cysteine Endopeptidases/chemistry , Cysteine Proteinase Inhibitors/chemical synthesis , Cysteine Proteinase Inhibitors/pharmacokinetics , Cysteine Proteinase Inhibitors/toxicity , Dipeptides/chemical synthesis , Dipeptides/pharmacokinetics , Dipeptides/toxicity , Drug Design , Female , Humans , Leishmania donovani/drug effects , Ligands , Mice , Microsomes, Liver/metabolism , Molecular Structure , Nitriles/chemical synthesis , Nitriles/pharmacokinetics , Nitriles/toxicity , Plasmodium falciparum/drug effects , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/chemistry , Rats , Structure-Activity Relationship , Swine , Triazoles/chemical synthesis , Triazoles/pharmacokinetics , Triazoles/toxicity , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/pharmacokinetics , Trypanocidal Agents/toxicity , Trypanosoma brucei rhodesiense/drug effects , Trypanosoma cruzi/drug effects
9.
Neurotox Res ; 34(1): 16-31, 2018 07.
Article in English | MEDLINE | ID: mdl-29218504

ABSTRACT

Parkinson's disease (PD) is conventionally seen as resulting from single-system neurodegeneration affecting nigrostriatal dopaminergic neurons. However, accumulating evidence indicates multi-system degeneration and neurotransmitter deficiencies, including cholinergic neurons which degenerate in a brainstem nucleus, the pedunculopontine nucleus (PPN), resulting in motor and cognitive impairments. The neuropeptide galanin can inhibit cholinergic transmission, while being upregulated in degenerating brain regions associated with cognitive decline. Here we determined the temporal-spatial profile of progressive expression of endogenous galanin within degenerating cholinergic neurons, across the rostro-caudal axis of the PPN, by utilizing the lactacystin-induced rat model of PD. First, we show progressive neuronal death affecting nigral dopaminergic and PPN cholinergic neurons, reflecting that seen in PD patients, to facilitate use of this model for assessing the therapeutic potential of bioactive peptides. Next, stereological analyses of the lesioned brain hemisphere found that the number of PPN cholinergic neurons expressing galanin increased by 11%, compared to sham-lesioned controls, and increasing by a further 5% as the neurodegenerative process evolved. Galanin upregulation within cholinergic PPN neurons was most prevalent closest to the intra-nigral lesion site, suggesting that galanin upregulation in such neurons adapt intrinsically to neurodegeneration, to possibly neuroprotect. This is the first report on the extent and pattern of galanin expression in cholinergic neurons across distinct PPN subregions in both the intact rat CNS and lactacystin-lesioned rats. The findings pave the way for future work to target galanin signaling in the PPN, to determine the extent to which upregulated galanin expression could offer a viable treatment strategy for ameliorating PD symptoms associated with cholinergic degeneration.


Subject(s)
Acetylcysteine/analogs & derivatives , Choline O-Acetyltransferase/metabolism , Cysteine Proteinase Inhibitors/toxicity , Galanin/metabolism , Neurons/pathology , Parkinson Disease , Pedunculopontine Tegmental Nucleus/pathology , Acetylcysteine/toxicity , Analysis of Variance , Animals , Disease Models, Animal , Male , Neurons/metabolism , Parkinson Disease/etiology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Rats , Rats, Sprague-Dawley , Substantia Nigra/drug effects , Substantia Nigra/pathology , Time Factors , Tyrosine 3-Monooxygenase/metabolism
10.
Naunyn Schmiedebergs Arch Pharmacol ; 391(1): 71-82, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29085973

ABSTRACT

The cathepsin B inhibitor benzyloxycarbonyl-phenylalanine-alanine-chloromethyl ketone (z-FA-CMK) was recently found to induce apoptosis at low concentrations in Jurkat T cells, while at higher concentrations, the cells die of necrosis. In the present study, we showed that z-FA-CMK readily depletes intracellular glutathione (GSH) with a concomitant increase in reactive oxygen species (ROS) generation. The toxicity of z-FA-CMK in Jurkat T cells was completely abrogated by N-acetylcysteine (NAC), suggesting that the toxicity mediated by z-FA-CMK is due to oxidative stress. We found that L-buthionine sulfoximine (BSO) which depletes intracellular GSH through the inhibition of GSH biosynthesis in Jurkat T cells did not promote ROS increase or induce cell death. However, NAC was still able to block z-FA-CMK toxicity in Jurkat T cells in the presence of BSO, indicating that the protective effect of NAC does not involve GSH biosynthesis. This is further corroborated by the protective effect of the non-metabolically active D-cysteine on z-FA-CMK toxicity. Furthermore, in BSO-treated cells, z-FA-CMK-induced ROS increased which remains unchanged, suggesting that the depletion of GSH and increase in ROS generation mediated by z-FA-CMK may be two separate events. Collectively, our results demonstrated that z-FA-CMK toxicity is mediated by oxidative stress through the increase in ROS generation.


Subject(s)
Cathepsin B/antagonists & inhibitors , Cathepsin B/metabolism , Cysteine Proteinase Inhibitors/toxicity , Leukemia, T-Cell/metabolism , Oxidative Stress/drug effects , Cell Death/drug effects , Cell Death/physiology , Dose-Response Relationship, Drug , Humans , Jurkat Cells , Oxidative Stress/physiology , Reactive Oxygen Species
11.
J Med Chem ; 60(16): 6911-6923, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28763614

ABSTRACT

This paper describes the development of a class of peptide-based inhibitors as novel antitrypanosomal and antimalarial agents. The inhibitors are based on a characteristic peptide sequence for the inhibition of the cysteine proteases rhodesain of Trypanosoma brucei rhodesiense and falcipain-2 of Plasmodium falciparum. We exploited the reactivity of novel unsaturated electrophilic functions such as vinyl-sulfones, -ketones, -esters, and -nitriles. The Michael acceptors inhibited both rhodesain and falcipain-2, at nanomolar and micromolar levels, respectively. In particular, the vinyl ketone 3b has emerged as a potent rhodesain inhibitor (k2nd = 67 × 106 M-1 min-1), endowed with a picomolar binding affinity (Ki = 38 pM), coupled with a single-digit micromolar activity against Trypanosoma brucei brucei (EC50 = 2.97 µM), thus being considered as a novel lead compound for the discovery of novel effective antitrypanosomal agents.


Subject(s)
Antimalarials/pharmacology , Carbamates/pharmacology , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Phenylalanine/analogs & derivatives , Trypanocidal Agents/pharmacology , Antimalarials/chemical synthesis , Antimalarials/toxicity , Carbamates/chemical synthesis , Carbamates/toxicity , Cathepsin L/metabolism , Cysteine Proteinase Inhibitors/chemical synthesis , Cysteine Proteinase Inhibitors/toxicity , Dipeptides/chemical synthesis , Dipeptides/toxicity , HeLa Cells , Humans , Hydrogen Bonding , Malaria/drug therapy , Molecular Docking Simulation , Molecular Dynamics Simulation , Neglected Diseases/drug therapy , Phenylalanine/chemical synthesis , Phenylalanine/pharmacology , Phenylalanine/toxicity , Plasmodium falciparum/drug effects , Stereoisomerism , Structure-Activity Relationship , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/toxicity , Trypanosoma brucei brucei/drug effects , Trypanosomiasis, African/drug therapy
12.
Exp Brain Res ; 235(7): 2189-2202, 2017 07.
Article in English | MEDLINE | ID: mdl-28439627

ABSTRACT

Proteinaceous inclusions, called Lewy bodies, are used as a pathological hallmark for Parkinson's disease (PD). Lewy bodies contain insoluble α-synuclein (aSyn) and many other ubiquitinated proteins, suggesting a role for protein degradation system failure in the PD pathogenesis. Indeed, proteasomal dysfunction has been linked to PD but commonly used in vivo toxin models, such as 6-OHDA or MPTP, do not have a significant effect on the proteasomal system or protein aggregation. Therefore, we wanted to study the characteristics of a proteasomal inhibitor, lactacystin, as a PD model on young and adult mice. To study this, we performed stereotactic microinjection of lactacystin above the substantia nigra pars compacta in young (2 month old) and adult (12-14 month old) C57Bl/6 mice. Motor behavior was measured by locomotor activity and cylinder tests, and the markers of neuroinflammation, aSyn, and dopaminergic system were assessed by immunohistochemistry and HPLC. We found that lactacystin induced a Parkinson's disease-like motor phenotype 5-7 days after injection in young and adult mice, and this was associated with widespread neuroinflammation based on glial cell markers, aSyn accumulation in substantia nigra, striatal dopamine decrease, and loss of dopaminergic cell bodies in the substantia nigra and terminals in the striatum. When comparing young and adult mice, adult mice were more sensitive for dopaminergic degeneration after lactacystin injection that further supports the use of adult mice instead of young when modeling neurodegeneration. Our data showed that lactacystin is useful in modeling various aspects of Parkinson's disease, and taken together, our findings emphasize the role of a protein degradation deficit in Parkinson's disease pathology, and support the use of proteasomal inhibitors as Parkinson's disease models.


Subject(s)
Acetylcysteine/analogs & derivatives , Cysteine Proteinase Inhibitors/toxicity , Neuroglia/drug effects , Parkinson Disease/etiology , Parkinson Disease/pathology , Substantia Nigra/drug effects , Acetylcysteine/toxicity , Age Factors , Animals , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Forelimb/physiopathology , Glial Fibrillary Acidic Protein/metabolism , Glutamate Decarboxylase/metabolism , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Microinjections , Neurotransmitter Agents/metabolism , Psychomotor Performance/drug effects , Synucleins/metabolism , Tyrosine 3-Monooxygenase/metabolism
13.
Exp Neurol ; 290: 15-28, 2017 04.
Article in English | MEDLINE | ID: mdl-28024798

ABSTRACT

Zonisamide (ZNS), an anticonvulsant drug exhibiting symptomatic effects in Parkinson's disease (PD), was recently reported to exert neuroprotection in rodent models. One of the proposed neuroprotective mechanisms involves increased protein expression of xCT, the specific subunit of the cystine/glutamate antiporter system xc-, inducing glutathione (GSH) synthesis. Here, we investigated the outcome of ZNS treatment in a mouse model of PD based on intranigral proteasome inhibition, and whether the observed effects would be mediated by system xc-. The proteasome inhibitor lactacystin (LAC) was administered intranigrally to male C57BL/6J mice receiving repeated intraperitoneal injections of either ZNS 30mgkg-1 or vehicle. Drug administration was initiated three days prior to stereotaxic LAC injection and was maintained until six days post-surgery. One week after lesion, mice were behaviorally assessed and investigated in terms of nigrostriatal neurodegeneration and molecular changes at the level of the basal ganglia, including expression levels of xCT. ZNS reduced the loss of nigral dopaminergic neurons following LAC injection and the degree of sensorimotor impairment. ZNS failed, however, to modulate xCT expression in basal ganglia of lesioned mice. In a separate set of experiments, the impact of ZNS treatment on system xc- was investigated in control conditions in vivo as well as in vitro. Similarly, ZNS did not influence xCT or glutathione levels in naive male C57BL/6J mice, nor did it alter system xc- activity or glutathione content in vitro. Taken together, these results demonstrate that ZNS treatment provides neuroprotection and behavioral improvement in a PD mouse model based on proteasome inhibition via system xc- independent mechanisms.


Subject(s)
Acetylcysteine/analogs & derivatives , Amino Acid Transport System y+/drug effects , Cysteine Proteinase Inhibitors/toxicity , Isoxazoles/pharmacology , Neuroprotective Agents/pharmacology , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/prevention & control , Acetylcysteine/administration & dosage , Acetylcysteine/antagonists & inhibitors , Acetylcysteine/toxicity , Animals , Basal Ganglia/drug effects , Basal Ganglia/metabolism , Basal Ganglia/pathology , Behavior, Animal/drug effects , Cysteine Proteinase Inhibitors/administration & dosage , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Glutathione/metabolism , Male , Mice , Mice, Inbred C57BL , Microinjections , Motor Activity/drug effects , Parkinson Disease, Secondary/psychology , Postural Balance/drug effects , Stereotaxic Techniques , Substantia Nigra , Zonisamide
14.
Bioorg Med Chem ; 24(18): 4228-4240, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27475533

ABSTRACT

In this study, we designed and synthesized a series of thiophen-2-iminothiazolidine derivatives from thiophen-2-thioureic with good anti-Trypanosoma cruzi activity. Several of the final compounds displayed remarkable trypanocidal activity. The ability of the new compounds to inhibit the activity of the enzyme cruzain, the major cysteine protease of T. cruzi, was also explored. The compounds 3b, 4b, 8b and 8c were the most active derivatives against amastigote form, with significant IC50 values between 9.7 and 6.03µM. The 8c derivative showed the highest potency against cruzain (IC50=2.4µM). Molecular docking study showed that this compound can interact with subsites S1 and S2 simultaneously, and the negative values for the theoretical energy binding (Eb=-7.39kcal·mol(-1)) indicates interaction (via dipole-dipole) between the hybridized sulfur sp(3) atom at the thiazolidine ring and Gly66. Finally, the results suggest that the thiophen-2-iminothiazolidines synthesized are important lead compounds for the continuing battle against Chagas disease.


Subject(s)
Thiazolidines/pharmacology , Thiophenes/pharmacology , Trypanocidal Agents/pharmacology , Trypanosoma cruzi/drug effects , Animals , Cell Line , Cysteine Endopeptidases , Cysteine Proteinase Inhibitors/chemical synthesis , Cysteine Proteinase Inhibitors/pharmacology , Cysteine Proteinase Inhibitors/toxicity , Glycine/chemistry , Mice , Molecular Docking Simulation , Octoxynol , Protozoan Proteins/antagonists & inhibitors , Thiazolidines/chemical synthesis , Thiazolidines/toxicity , Thiophenes/chemical synthesis , Thiophenes/toxicity , Thiourea/analogs & derivatives , Thiourea/chemical synthesis , Thiourea/pharmacology , Thiourea/toxicity , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/toxicity
15.
Mol Neurobiol ; 53(7): 4939-60, 2016 09.
Article in English | MEDLINE | ID: mdl-26374549

ABSTRACT

Astrocytes are one of the major cell types to combat cellular stress and protect neighboring neurons from injury. In order to fulfill this important role, astrocytes must sense and respond to toxic stimuli, perhaps including stimuli that are severely stressful and kill some of the astrocytes. The present study demonstrates that primary astrocytes that managed to survive severe proteotoxic stress were protected against subsequent challenges. These findings suggest that the phenomenon of preconditioning or tolerance can be extended from mild to severe stress for this cell type. Astrocytic stress adaptation lasted at least 96 h, the longest interval tested. Heat shock protein 70 (Hsp70) was raised in stressed astrocytes, but inhibition of neither Hsp70 nor Hsp32 activity abolished their resistance against a second proteotoxic challenge. Only inhibition of glutathione synthesis abolished astrocytic stress adaptation, consistent with our previous report. Primary neurons were plated upon previously stressed astrocytes, and the cocultures were then exposed to another proteotoxic challenge. Severely stressed astrocytes were still able to protect neighboring neurons against this injury, and the protection was unexpectedly independent of glutathione synthesis. Stressed astrocytes were even able to protect neurons after simultaneous application of proteasome and Hsp70 inhibitors, which otherwise elicited synergistic, severe loss of neurons when applied together. Astrocyte-induced neuroprotection against proteotoxicity was not elicited with astrocyte-conditioned media, suggesting that physical cell-to-cell contacts may be essential. These findings suggest that astrocytes may adapt to severe stress so that they can continue to protect neighboring cell types from profound injury.


Subject(s)
Astrocytes/physiology , Neurons/physiology , Oxidative Stress/physiology , Animals , Animals, Newborn , Astrocytes/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Coculture Techniques , Cysteine Proteinase Inhibitors/toxicity , HSP70 Heat-Shock Proteins/metabolism , Neurons/drug effects , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley
16.
Parasitol Int ; 65(1): 20-22, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26384965

ABSTRACT

Hypervalent organotellurium compounds (organotelluranes) have shown several promising applications, including their use as potent and selective cysteine protease inhibitors and antiprotozoal agents. Here, we report the antimalarial activities of three organotellurane derivatives (RF05, RF07 and RF19) in two Plasmodium falciparum strains (CQS 3D7 and CQR W2), which demonstrated significant decreases in parasitemia in vitro. The inhibition of intracellular P. falciparum proteases by RF05, RF07 and RF19 was determined and the IC50 values were 3.7±1.0µM, 1.1±0.2µM and 0.2±0.01µM, respectively. Using an assay performed in the presence of the ER Ca(2+)-ATPase inhibitor we showed that the main enzymatic targets were cysteine proteases stimulated by calcium (calpains). None of the compounds tested caused haemolysis or a significant decrease in endothelial cell viability in the concentration range used for the inhibition assay. Taken together, the results suggest promising compounds for the development of antimalarial drugs.


Subject(s)
Antimalarials/pharmacology , Calpain/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Organometallic Compounds/pharmacology , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Tellurium/pharmacology , Antimalarials/toxicity , Calcium/metabolism , Cell Survival/drug effects , Cysteine Proteinase Inhibitors/toxicity , Drug Discovery , Erythrocytes/drug effects , Erythrocytes/parasitology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/parasitology , Humans , Inhibitory Concentration 50 , Malaria, Falciparum/drug therapy , Organometallic Compounds/toxicity , Tellurium/toxicity
17.
J Neural Transm (Vienna) ; 122(12): 1645-57, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26253900

ABSTRACT

The leucine-rich repeat kinase 2 (LRRK2) mutation G2019S is one of the most common genetic causes in Parkinson's disease (PD). The penetrance of G2019S LRRK2 is incomplete and is age-dependent, therefore, it has been speculated that environmental toxins and aging could contribute to G2019S LRRK2-related PD pathogenesis. To prove this speculation, we performed a longitudinal investigation in mice bearing G2019S LRRK2 mutation. BAC G2019S LRRK2 transgenic (Tg) mice and their wildtype (Wt) littermates were treated with lactacystin, a specific proteasome inhibitor. The susceptibilities of mice to lactacystin-induced nigrostriatal dopaminergic (DAergic) degeneration were evaluated, at 5 and 12 months of age. We found that lactacystin treatment caused a greater decline of striatal DA content in the Tg mice at either 5 or 12 months of age than their age-matched Wt littermates. Moreover, the lactacystin-treated Tg or Wt mice at 12 months of age lose much more nigral tyrosine hydroxylase (TH)-positive neurons than the mice at 5 months of age, indicating an age-associated DAergic neurotoxicity. Additionally, stereotactic injection of lactacystin induced a dramatic increase of activated microglia in substantia nigra of mice at 12 months of age, compared with mice at 5 months of age. In summary, our study suggests that expression of the G2019S mutation in the mouse LRRK2 gene confers an age-associated high susceptibility to proteasome inhibition-induced nigrostriatal DAergic degeneration.


Subject(s)
Aging/physiology , Corpus Striatum/physiopathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/physiopathology , Protein Serine-Threonine Kinases/metabolism , Substantia Nigra/physiopathology , Acetylcysteine/analogs & derivatives , Acetylcysteine/toxicity , Aging/genetics , Animals , Corpus Striatum/pathology , Cysteine Proteinase Inhibitors/toxicity , Disease Models, Animal , Dopaminergic Neurons/pathology , Dopaminergic Neurons/physiology , Genetic Predisposition to Disease , Mice, Transgenic , Microglia/pathology , Microglia/physiology , Motor Activity/physiology , Mutation , Neurodegenerative Diseases/pathology , Protein Serine-Threonine Kinases/genetics , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/metabolism
18.
Rev. bras. ginecol. obstet ; 37(1): 42-51, 01/2015. tab
Article in English | LILACS | ID: lil-732870

ABSTRACT

Gestational trophoblastic neoplasia (GTN) is the term to describe a set of malignant placental diseases, including invasive mole, choriocarcinoma, placental site trophoblastic tumor and epithelioid trophoblastic tumor. Both invasive mole and choriocarcinoma respond well to chemotherapy, and cure rates are greater than 90%. Since the advent of chemotherapy, low-risk GTN has been treated with a single agent, usually methotrexate or actinomycin D. Cases of high-risk GTN, however, should be treated with multiagent chemotherapy, and the regimen usually selected is EMA-CO, which combines etoposide, methotrexate, actinomycin D, cyclophosphamide and vincristine. This study reviews the literature about GTN to discuss current knowledge about its diagnosis and treatment.


Neoplasia trofoblástica gestacional (NTG) é o termo que descreve o conjunto de anomalias malignas da placenta, incluindo a mola invasora, coriocarcinoma, tumor trofoblástico do sítio placentário e tumor trofoblástico epitelióide. Ambos a mola invasora e o coriocarcinoma respondem bem à quimioterapia, com taxas de cura superiores a 90%. Desde o advento da quimioterapia, NTG de baixo risco tem sido tratada com monoquimioterapia, pelo geral methotrexate ou actinomicina-D. Casos de NTG de alto risco, contudo, devem ser tratados com poliquimioterapia, e o regime usualmente escolhido é o EMA-CO que combina etoposide, methotrexate, actinomicina-D, ciclofosfamida e vincristina. Esse estudo revê a literatura sobre NTG a fim de discutir os conhecimentos atuais sobre seu diagnóstico e tratamento.


Subject(s)
Animals , Male , Rats , Cathepsins/analysis , Cystatins/analysis , Cysteine Proteinase Inhibitors/metabolism , Endopeptidases , Leucine/analogs & derivatives , Osteoclasts/chemistry , Osteoclasts/enzymology , Salivary Proteins and Peptides/analysis , Bone Matrix/chemistry , Bone Matrix/enzymology , Cathepsin L , Cysteine Endopeptidases , Cathepsins/antagonists & inhibitors , Cathepsins/metabolism , Cystatins/metabolism , Cysteine Proteinase Inhibitors/toxicity , Leucine/metabolism , Leucine/toxicity , Lysosomes/enzymology , Microscopy, Immunoelectron , Osteoclasts/drug effects , Osteoclasts/ultrastructure , Rats, Wistar , Salivary Cystatins
19.
Brain Struct Funct ; 220(1): 479-500, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24292256

ABSTRACT

A rostral brainstem structure, the pedunculopontine nucleus (PPN), is severely affected by Parkinson's disease (PD) pathology and is regarded a promising target for therapeutic deep-brain stimulation (DBS). However, understanding the PPN's role in PD and assessing the potential of DBS are hampered by the lack of a suitable model of PPN degeneration. Rats were rendered Parkinsonian through a unilateral substantia nigra pars compacta (SNpc) stereotaxic injection of the proteasome inhibitor Lactacystin, to investigate whether the lesion's pathological effects spread to impact the integrity of PPN cholinergic neurons which are affected in PD. At 5 weeks post-surgery, stereological analysis revealed that the lesion caused a 48 % loss of dopaminergic SNpc neurons and a 61 % loss of PPN cholinergic neurons, accompanied by substantial somatic hypotrophy in the remaining cholinergic neurons. Magnetic resonance imaging revealed T2 signal hyper-/hypointensity in the PPN of the injected hemisphere, respectively at weeks 3 and 5 post-lesion. Moreover, isolated PPN cholinergic neurons revealed no significant alterations in key autophagy mRNA levels, suggesting that autophagy-related mechanisms fail to protect the PPN against Lactacystin-induced cellular changes. Hence, the current results suggest that the Lactacystin PD model offers a suitable model for investigating the role of the PPN in PD.


Subject(s)
Cholinergic Neurons/pathology , Disease Models, Animal , Nerve Degeneration/etiology , Parkinson Disease/complications , Pedunculopontine Tegmental Nucleus/pathology , Acetylcysteine/analogs & derivatives , Acetylcysteine/toxicity , Analysis of Variance , Animals , Antibodies, Monoclonal/metabolism , Calcium-Binding Proteins/metabolism , Cell Count , Choline O-Acetyltransferase/metabolism , Cysteine Proteinase Inhibitors/toxicity , Forelimb/physiopathology , Functional Laterality , Image Processing, Computer-Assisted , Laser Capture Microdissection , Magnetic Resonance Imaging , Male , Membrane Proteins/metabolism , Motor Activity , Muscle Proteins/metabolism , Parkinson Disease/etiology , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase/metabolism
20.
Cardiovasc Toxicol ; 14(3): 275-87, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24615591

ABSTRACT

Lipid peroxidation (LP), induced by oxidative stress, is associated with degenerative processes. 4-Hydroxy-2-nonenal (HNE), a highly reactive diffusible product of LP, is considered by-product and mediator of oxidative stress. Its level increases under pathological conditions such as cardiovascular diseases. In this study, we partially characterized the mechanisms of HNE-mediated cytotoxicity in cardiomyocytes. After establishing that pathophysiological doses of HNE trigger cell death dependent on the incubation time and dose of HNE (LD50 = 4.4 µM), we tackled the mechanisms that underlie the cell death induced by HNE. Our results indicate that HNE rapidly increases intracellular Ca(2+); it also increases the rate of reactive oxygen species generation and causes a loss of mitochondrial membrane potential (ΔΨm) as well as a decrease in the ATP and GSH levels. Such alterations result in the activation of caspase-3 and DNA breakdown, both characteristic features of apoptotic cell death, as well as disruption of the cytoskeleton. Moreover, the nucleophilic compounds N-acetyl-cysteine and ß-mercapto-propionyl-glycine, and the synthetic antioxidant Trolox exert a potent antioxidant action against HNE damage; this suggests its use as effective compounds in order to reduce the damage occurred as consequence of cardiovascular disorders in which oxidative stress and hence LP take place.


Subject(s)
Aldehydes/toxicity , Apoptosis/drug effects , Cysteine Proteinase Inhibitors/toxicity , Myocytes, Cardiac/pathology , Acetylcysteine/pharmacology , Animals , Animals, Newborn , Antioxidants/pharmacology , Apoptosis/physiology , Calcium/metabolism , Cardiotoxicity/pathology , Cells, Cultured , Chromans/pharmacology , Cytoskeleton/pathology , Flow Cytometry , Lipid Peroxidation , Membrane Potential, Mitochondrial/drug effects , Oxidative Stress , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...