Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Type of study
Language
Publication year range
1.
Brain Behav Immun ; 83: 248-259, 2020 01.
Article in English | MEDLINE | ID: mdl-31669344

ABSTRACT

Chemotherapy-induced neuropathic pain is a dose-limiting side effect of many cancer therapies due to their propensity to accumulate in peripheral nerves, which is facilitated by the permeability of the blood-nerve barrier. Preclinically, the chemotherapy agent vincristine (VCR) activates endothelial cells in the murine peripheral nervous system and in doing so allows the infiltration of monocytes into nerve tissue where they orchestrate the development of VCR-induced nociceptive hypersensitivity. In this study we demonstrate that VCR also activates endothelial cells in the murine central nervous system, increases paracellular permeability and decreases trans endothelial resistance. In in vivo imaging studies in mice, VCR administration results in trafficking of inflammatory monocytes through the endothelium. Indeed, VCR treatment affects the integrity of the blood-spinal cord-barrier as indicated by Evans Blue extravasation, disrupts tight junction coupling and is accompanied by the presence of monocytes in the spinal cord. Such inflammatory monocytes (Iba-1+ CCR2+ Ly6C+ TMEM119- cells) that infiltrate the spinal cord also express the pro-nociceptive cysteine protease Cathepsin S. Systemic treatment with a CNS-penetrant, but not a peripherally-restricted, inhibitor of Cathepsin S prevents the development of VCR-induced hypersensitivity, suggesting that infiltrating monocytes play a functional role in sensitising spinal cord nociceptive neurons. Our findings guide us towards a better understanding of central mechanisms of pain associated with VCR treatment and thus pave the way for the development of innovative antinociceptive strategies.


Subject(s)
Capillary Permeability , Drug-Related Side Effects and Adverse Reactions , Neuralgia/physiopathology , Spinal Cord/blood supply , Animals , Endothelial Cells/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Neuralgia/chemically induced
2.
Neurobiol Pain ; 6: 100032, 2019.
Article in English | MEDLINE | ID: mdl-31223140

ABSTRACT

Chronic itch is a debilitating condition characterised by excessive scratching and is a symptom frequently reported in skin diseases such as atopic dermatitis. It has been proposed that release of the cysteine protease Cathepsin S (CatS) from skin keratinocytes or immune cells resident in or infiltrating the skin could act as a pruritogen in chronic itch conditions. CatS is known to activate protease-activated receptor 2 (PAR2). We therefore hypothesised that enzymatic activation of neuronally expressed PAR2 by CatS was responsible for activation of sensory neurons and transmission of itch signals. Intradermally-injected human recombinant (hr)-CatS or the PAR2 agonist, SLIGRL-NH2 behaved as pruritogens by causing scratching behaviour in mice. Hr-CatS-induced scratching behaviour was prevented by CatS inhibitors and PAR2 antagonists and reduced by 50% in TRPV1-/- mice compared with wild-type mice, whilst no significant reduction in scratching behaviour was observed in TRPA1-/- mice. Cultured dorsal root ganglion (DRG) cells showed an increase in [Ca2+]i following incubation with hr-CatS, and the percentage of neurons that responded to hr-CatS decreased in the presence of a PAR2 antagonist or in cultures of neurons from TRPV1-/- mice. Taken together, our results indicate CatS acts as a pruritogen via PAR2 activation in TRPV1-expressing sensory neurons.

3.
J Pharmacol Exp Ther ; 358(3): 387-96, 2016 09.
Article in English | MEDLINE | ID: mdl-27335437

ABSTRACT

Cathepsin S inhibitors attenuate mechanical allodynia in preclinical neuropathic pain models. The current study evaluated the effects when combining the selective cathepsin S inhibitor MIV-247 with gabapentin or pregabalin in a mouse model of neuropathic pain. Mice were rendered neuropathic by partial sciatic nerve ligation. MIV-247, gabapentin, or pregabalin were administered alone or in combination via oral gavage. Mechanical allodynia was assessed using von Frey hairs. Neurobehavioral side effects were evaluated by assessing beam walking. MIV-247, gabapentin, and pregabalin concentrations in various tissues were measured. Oral administration of MIV-247 (100-200 µmol/kg) dose-dependently attenuated mechanical allodynia by up to approximately 50% reversal when given as a single dose or when given twice daily for 5 days. No behavioral deficits were observed at any dose of MIV-247 tested. Gabapentin (58-350 µmol/kg) and pregabalin (63-377 µmol/kg) also inhibited mechanical allodynia with virtually complete reversal at the highest doses tested. The minimum effective dose of MIV-247 (100 µmol/kg) in combination with the minimum effective dose of pregabalin (75 µmol/kg) or gabapentin (146 µmol/kg) resulted in enhanced antiallodynic efficacy without augmenting side effects. A subeffective dose of MIV-247 (50 µmol/kg) in combination with a subeffective dose of pregabalin (38 µmol/kg) or gabapentin (73 µmol/kg) also resulted in substantial efficacy. Plasma levels of MIV-247, gabapentin, and pregabalin were similar when given in combination as to when given alone. Cathepsin S inhibition with MIV-247 exerts significant antiallodynic efficacy alone, and also enhances the effect of gabapentin and pregabalin without increasing side effects or inducing pharmacokinetic interactions.


Subject(s)
Amines/pharmacology , Cathepsins/antagonists & inhibitors , Cyclohexanecarboxylic Acids/pharmacology , Dipeptides/pharmacology , Hyperalgesia/drug therapy , Neuralgia/drug therapy , Pregabalin/pharmacology , Protease Inhibitors/pharmacology , gamma-Aminobutyric Acid/pharmacology , Animals , Behavior, Animal/drug effects , Dipeptides/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Gabapentin , Humans , Hyperalgesia/enzymology , Male , Mice , Neuralgia/enzymology , Protease Inhibitors/therapeutic use
4.
Eur J Pharmacol ; 613(1-3): 155-62, 2009 Jun 24.
Article in English | MEDLINE | ID: mdl-19358841

ABSTRACT

Cathepsin K (EC 3.4.22.38) is expressed by osteoclasts and synovial fibroblasts and its proteolytic activity is hypothesized to play a role in the pathology of rheumatoid arthritis. This study explored the effects of the cathepsin K inhibitor N-(1-{[(Cyanomethyl)amino]carbonyl}cyclohexyl)-4-[2-(4-methylpiperazin-1-yl)-1,3-thiazol-4-yl]benzamide (L-006235) in murine collagen-induced arthritis. L-006235 is a potent inhibitor of recombinant human and murine cathepsin K, enzymes (K(i):0.073 nM and IC(50): 2.4 nM, respectively) and at the cellular level in human osteoclasts (IC(50): 28 nM) with ~1000-fold selectivity against cathepsin S. L-006235 did not result in splenic invariant chain p10 accumulation, a specific marker of cathepsin S inhibition. L-006235 was dosed daily (25 mg/kg, p.o.), either prophylactically (days 0-42) or therapeutically (14 days post onset of disease) to DBA/1J mice subjected to collagen-induced arthritis. Disease severity was scored during the course of the study. Histological evaluation of cartilage and bone degradation together with related biomarkers namely, deoxypyridinoline, cartilage oligomeric matrix protein and C-terminal telopeptide degradation product of type I collagen (CTX-I) were analyzed after the study. After prophylactic or therapeutic administration, L-006235 significantly reduced biomarkers reflecting bone and cartilage degradation. Pathological changes at the histological level were significantly reduced after prophylactic treatment (P<0.01), but not after therapeutic treatment. Prophylactic treatment with L-006235 delayed disease onset (P<0.01) and reduced the disease severity score (P<0.05). Inhibition of cathepsin K activity exerts beneficial effects on collagen-induced arthritis in mice and thus warrants further investigation as a therapeutic intervention in human rheumatoid arthritis.


Subject(s)
Arthritis, Experimental/drug therapy , Bone and Bones/drug effects , Cartilage/drug effects , Cathepsins/antagonists & inhibitors , Collagen/adverse effects , Inflammation/drug therapy , Protease Inhibitors/pharmacology , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/complications , Arthritis, Experimental/prevention & control , Benzamides/pharmacokinetics , Benzamides/pharmacology , Benzamides/therapeutic use , Biomarkers/metabolism , Bone Resorption/drug therapy , Bone Resorption/etiology , Bone and Bones/metabolism , Bone and Bones/pathology , Cartilage/metabolism , Cartilage/pathology , Cathepsin K , Cathepsins/isolation & purification , Female , Humans , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Mice , Protease Inhibitors/pharmacokinetics , Protease Inhibitors/therapeutic use , Thiazoles/pharmacokinetics , Thiazoles/pharmacology , Thiazoles/therapeutic use
5.
Bioorg Med Chem ; 17(3): 1307-24, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19124252

ABSTRACT

Highly potent and selective 4-amidofuran-3-one inhibitors of cathepsin S are described. The synthesis and structure-activity relationship of a series of inhibitors with a sulfonamide moiety in the P3 position is presented. Several members of the series show sub-nanomolar inhibition of the target enzyme as well as an excellent selectivity profile and good cellular potency. Molecular modeling of the most interesting inhibitors describes interactions in the extended S3 pocket and explains the observed selectivity towards cathepsin K.


Subject(s)
Cathepsins/antagonists & inhibitors , Furans/chemistry , Protease Inhibitors/chemistry , Sulfonamides/chemistry , Cathepsin K , Computer Simulation , Furans/chemical synthesis , Furans/pharmacology , Humans , Models, Molecular , Protease Inhibitors/chemical synthesis , Protease Inhibitors/pharmacology , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/pharmacology
6.
Biol Chem ; 384(8): 1239-46, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12974392

ABSTRACT

Mammalian asparaginyl endopeptidase (AEP) or legumain is a recently identified lysosomal cysteine protease belonging to clan CD. To date it has been shown to be involved in antigen presentation within class II MHC positive cells and in pro-protein processing. Further elucidation of the biological functions of the enzyme will require potent and selective inhibitors and thus we describe here new acyloxymethylketone inhibitors of AEP. The most potent of the series is 2,6-dimethyl-benzoic acid 3-benzyloxycarbonylamino-4-carbamoyl-2-oxo-butyl ester (MV026630) with a kobs/[I] value of 1.09 x 10(5) M(-1) s(-1). At low microM concentrations this compound is able to enter living cells and irreversibly inactivate AEP. We show that this results in inhibition of AEP autoactivation and in perturbation of the processing and presentation of T cell epitopes from both tetanus toxin and myelin basic protein.


Subject(s)
Benzoates/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Animals , Antigen Presentation/drug effects , Antigens, Bacterial/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/enzymology , B-Lymphocytes/immunology , Benzoates/chemistry , Catalysis , Cell Line, Tumor , Cell Membrane Permeability , Cysteine Proteinase Inhibitors/chemistry , Humans , Kinetics , Molecular Structure , Structure-Activity Relationship , Substrate Specificity , T-Lymphocytes/drug effects , T-Lymphocytes/enzymology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL