Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
2.
Basic Clin Pharmacol Toxicol ; 125(2): 89-99, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30916878

ABSTRACT

Proton pump inhibitors (PPIs) are prodrugs used in the treatment of peptic ulcer diseases. Once activated by acidic pH, the PPIs subsequently inhibit the secretion of gastric acid by covalently forming disulphide bonds with the SH groups of the parietal proton pump, that is the H+ /K+ -ATPase. Long-term use of PPIs has been associated with numerous adverse effects, including bone fractures. Considering the mechanism of activation, PPIs could also be active in acidic micro-environments such as in lysosomes, tumours and bone resorption sites. We suggested that the SH group in the active site of cysteine proteases could be susceptible for inhibition by PPIs. In this study, the inhibition by lansoprazole was shown on the cysteine proteases legumain and cathepsin B by incubating purified proteases or cell lysates with lansoprazole at different concentrations and pH conditions. The mechanism of legumain inhibition was shown to be a direct interaction of lansoprazole with the SH group in the active site, and thus blocking binding of the legumain-selective activity-based probe MP-L01. Lansoprazole was also shown to inhibit both legumain and cathepsin B in various cell models like HEK293, monoclonal legumain over-expressing HEK293 cells (M38L) and RAW264.7 macrophages, but not in human bone marrow-derived skeletal (mesenchymal) stem cells (hBMSC-TERT). During hBMSC-TERT differentiation to osteoblasts, lansoprazole inhibited legumain secretion, alkaline phosphatase activity, but had no effects on in vitro mineralization capacity. In conclusion, lansoprazole acts as a direct covalent inhibitor of cysteine proteases via disulphide bonds with the SH group in the protease active site. Such inhibition of cysteine proteases could explain some of the off-target effects of PPIs.


Subject(s)
Cysteine Proteinase Inhibitors/adverse effects , Lansoprazole/adverse effects , Proton Pump Inhibitors/adverse effects , Animals , Calcification, Physiologic/drug effects , Catalytic Domain/drug effects , Cathepsin B/antagonists & inhibitors , Cell Differentiation/drug effects , Cysteine Endopeptidases/metabolism , Fractures, Bone/chemically induced , HEK293 Cells , Humans , Mesenchymal Stem Cells , Mice , Osteoblasts/drug effects , Osteoblasts/physiology , Peptic Ulcer/drug therapy , Protein Binding , RAW 264.7 Cells
3.
Br J Clin Pharmacol ; 85(6): 1072-1083, 2019 06.
Article in English | MEDLINE | ID: mdl-30663085

ABSTRACT

Cathepsin K (CatK) is a cysteine protease abundantly expressed by osteoclasts and localized in the lysosomes and resorption lacunae of these cells. CatK is the principal enzyme responsible for the degradation of bone collagen. Odanacatib is a selective, reversible inhibitor of CatK at subnanomolar potency. The pharmacokinetics of odanacatib have been extensively studied and are similar in young healthy men, postmenopausal women and elderly men, and were qualitatively similar throughout Phase 1 development and in-patient studies. Following 3 weeks of 50 mg once weekly dosing the geometric mean area under the curve from 0 to 168 hours was 41.1 µM h, the concentration at 168 hours was 126 nM and the harmonic mean apparent terminal half-life was 84.8 hr. Odanacatib exposure increased in a less than dose proportional manner due to solubility limited absorption. It is estimated that approximately 70% of the absorbed dose of odanacatib is eliminated via metabolism, 20% is excreted as unchanged drug in the bile or faeces, and 10% is excreted as unchanged drug in the urine. The systemic clearance was low (approximately 13 mL/min). Odanacatib decreases the degradation of bone matrix proteins and reduces the efficiency of bone resorption with target engagement confirmed by a robust decrease in serum C-telopeptides of type 1 collagen (approximately 60%), urinary aminoterminal crosslinked telopeptides of type 1 collagen to creatinine ratio (approximately 50%) and total urine deoxypyridinoline/Cr (approximately 30%), with an increase in serum cross-linked carboxy-terminal telopeptide of type 1 collagen (approximately 55%). The 50-mg weekly dosing regimen evaluated in Phase 3 achieved near maximal reduction in bone resorption throughout the treatment period. The extensive clinical programme for odanacatib, together with more limited clinical experience with other CatK inhibitors (balicatib and ONO-5334), provides important insights into the clinical pharmacology of CatK inhibition and the potential role of CatK in bone turnover and mineral homeostasis. Key findings include the ability of this mechanism to: (i) provide sustained reductions in resorption markers, increases in bone mineral density, and demonstrated fracture risk reduction; (ii) be associated with relative formation-sparing effects such that sustained resorption reduction is achieved without accompanying meaningful reductions in bone formation; and (iii) lead to increases in osteoclast number as well as other osteoclast activity (including build-up of CatK enzyme), which may yield transient increases in resorption following treatment discontinuation and the potential for nonmonotonic responses at subtherapeutic doses.


Subject(s)
Biphenyl Compounds/therapeutic use , Bone Density Conservation Agents/therapeutic use , Bone Remodeling/drug effects , Bone and Bones/drug effects , Cathepsin K/antagonists & inhibitors , Cysteine Proteinase Inhibitors/therapeutic use , Osteoporosis/drug therapy , Animals , Biphenyl Compounds/adverse effects , Biphenyl Compounds/pharmacokinetics , Bone Density Conservation Agents/adverse effects , Bone Density Conservation Agents/pharmacokinetics , Bone and Bones/enzymology , Bone and Bones/physiopathology , Cathepsin K/metabolism , Cysteine Proteinase Inhibitors/adverse effects , Cysteine Proteinase Inhibitors/pharmacokinetics , Female , Humans , Male , Osteoporosis/enzymology , Osteoporosis/pathology , Signal Transduction , Translational Research, Biomedical , Treatment Outcome
4.
Clin Pharmacol Ther ; 104(6): 1155-1164, 2018 12.
Article in English | MEDLINE | ID: mdl-29484635

ABSTRACT

Neutrophil serine proteases (NSPs), such as neutrophil elastase (NE), are activated by dipeptidyl peptidase 1 (DPP1) during neutrophil maturation. High NSP levels can be detrimental, particularly in lung tissue, and inhibition of NSPs is therefore an interesting therapeutic opportunity in multiple lung diseases, including chronic obstructive pulmonary disease (COPD) and bronchiectasis. We conducted a randomized, placebo-controlled, first-in-human study to assess the safety, tolerability, pharmacokinetics, and pharmacodynamics of single and multiple oral doses of the DPP1 inhibitor AZD7986 in healthy subjects. Pharmacokinetic and pharmacodynamic data were analyzed using nonlinear mixed effects modeling and showed that AZD7986 inhibits whole blood NE activity in an exposure-dependent, indirect manner-consistent with in vitro and preclinical predictions. Several dose-dependent, possibly DPP1-related, nonserious skin findings were observed, but these were not considered to prevent further clinical development. Overall, the study results provided confidence to progress AZD7986 to phase II and supported selection of a clinically relevant dose.


Subject(s)
Benzoxazoles/administration & dosage , Cathepsin C/antagonists & inhibitors , Cysteine Proteinase Inhibitors/administration & dosage , Leukocyte Elastase/antagonists & inhibitors , Neutrophils/drug effects , Oxazepines/administration & dosage , Serine Proteinase Inhibitors/administration & dosage , Administration, Oral , Benzoxazoles/adverse effects , Benzoxazoles/pharmacokinetics , Cysteine Proteinase Inhibitors/adverse effects , Cysteine Proteinase Inhibitors/pharmacokinetics , Dose-Response Relationship, Drug , Drug Administration Schedule , Healthy Volunteers , Humans , Leukocyte Elastase/blood , Male , Models, Biological , Neutrophils/enzymology , Nonlinear Dynamics , Oxazepines/adverse effects , Oxazepines/pharmacokinetics , Serine Proteinase Inhibitors/pharmacokinetics
5.
Reprod Fertil Dev ; 29(6): 1107-1114, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27156102

ABSTRACT

In camelids, the development of assisted reproductive technologies is impaired by the viscous nature of the semen. The protease papain has shown promise in reducing viscosity, although its effect on sperm integrity is unknown. The present study determined the optimal papain concentration and exposure time to reduce seminal plasma viscosity and investigated the effect of papain and its inhibitor E-64 on sperm function and cryopreservation in alpacas. Papain (0.1mg mL-1, 20min, 37°C) eliminated alpaca semen viscosity while maintaining sperm motility, viability, acrosome integrity and DNA integrity. Furthermore E-64 (10 µM at 37°C for 5min after 20min papain) inhibited the papain without impairing sperm function. Cryopreserved, papain-treated alpaca spermatozoa exhibited higher total motility rates after chilling and 0 and 1h after thawing compared with control (untreated) samples. Papain treatment, followed by inhibition of papain with E-64, is effective in reducing alpaca seminal plasma viscosity without impairing sperm integrity and improves post-thaw motility rates of cryopreserved alpaca spermatozoa. The use of the combination of papain and E-64 to eliminate the viscous component of camelid semen may aid the development of assisted reproductive technologies in camelids.


Subject(s)
Cysteine Proteinase Inhibitors/pharmacology , Leucine/analogs & derivatives , Papain/metabolism , Semen/drug effects , Sperm Motility/drug effects , Acrosome Reaction/drug effects , Animals , Camelids, New World , Cell Survival/drug effects , Cryopreservation/veterinary , Cysteine Proteinase Inhibitors/adverse effects , DNA/drug effects , DNA/metabolism , Fructose/chemistry , Fructose/metabolism , In Situ Nick-End Labeling , Kinetics , Lactose/chemistry , Lactose/metabolism , Leucine/adverse effects , Leucine/pharmacology , Male , New South Wales , Papain/adverse effects , Papain/antagonists & inhibitors , Semen/physiology , Semen Analysis/veterinary , Semen Preservation/veterinary , Viscosity/drug effects
6.
Biochem Pharmacol ; 117: 10-9, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27106079

ABSTRACT

Osteoporosis is a metabolic bone disease that is characterized by heightened state of bone resorption accompanied by diminished bone formation, leading to a reduction of bone mineral density (BMD) and deterioration of bone quality, thus increasing the risk of developing fractures. Molecular insight into bone biology identified cathepsin K (CatK) as a novel therapeutic target. CatK is a lysosomal cysteine protease secreted by activated osteoclasts during bone resorption, whose primary substrate is type I collagen, the major component of organic bone matrix. Available anti-resorptive drugs affect osteoclast survival and influence both resorption and formation of bone. CatK inhibitors are distinct from the existing anti-resorptives as they only target the resorption process itself without impairing osteoclast differentiation and do not interfere with bone formation. An inhibitor of CatK, odanacatib, robustly increased both trabecular and cortical BMD in postmenopausal osteoporosis patients. The phase III fracture prevention trial with odanacatib ended early due to good efficacy and a favorable benefit/risk profile, thus, enhancing the opportunity for CatK as a pharmacological target for osteoporosis. So far, all the inhibitors that reached to the stage of clinical trial targeted active site of CatK to abrogate the entire proteolytic activity of the enzyme in addition to the desired blockage of excessive elastin and collagen degradation, and could thus pose safety concerns with long term use. Identification of selective exosite inhibitors that inhibit CatK's elastase and/or collagenase activity but do not affect the hydrolysis of other physiologically relevant substrates of CatK would be an improved strategy to inhibit this enzyme.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Bone and Bones/drug effects , Cathepsin K/antagonists & inhibitors , Cysteine Proteinase Inhibitors/therapeutic use , Drugs, Investigational/therapeutic use , Molecular Targeted Therapy , Osteoporosis, Postmenopausal/drug therapy , Aged , Animals , Biphenyl Compounds/adverse effects , Biphenyl Compounds/pharmacology , Biphenyl Compounds/therapeutic use , Bone Density Conservation Agents/adverse effects , Bone Density Conservation Agents/pharmacology , Bone and Bones/enzymology , Cathepsin K/metabolism , Cysteine Proteinase Inhibitors/adverse effects , Cysteine Proteinase Inhibitors/pharmacology , Drugs, Investigational/adverse effects , Drugs, Investigational/pharmacology , Female , Humans , Molecular Targeted Therapy/adverse effects , Osteoclasts/drug effects , Osteoclasts/enzymology , Osteoclasts/pathology , Osteoporosis, Postmenopausal/enzymology , Osteoporosis, Postmenopausal/pathology , Osteoporosis, Postmenopausal/physiopathology , Osteoporotic Fractures/etiology , Osteoporotic Fractures/pathology , Osteoporotic Fractures/prevention & control
7.
Blood ; 120(5): 947-59, 2012 Aug 02.
Article in English | MEDLINE | ID: mdl-22645181

ABSTRACT

Proteasome inhibition has emerged as an important therapeutic strategy in multiple myeloma (MM). Since the publication of the first phase 1 trials of bortezomib 10 years ago, this first-in-class proteasome inhibitor (PI) has contributed substantially to the observed improvement in survival in MM patients over the past decade. Although first approved as a single agent in the relapsed setting, bortezomib is now predominantly used in combination regimens. Furthermore, the standard twice-weekly schedule may be replaced by weekly infusion, especially when bortezomib is used as part of combination regimens in frontline therapy. Indeed, bortezomib is an established component of induction therapy for patients eligible or ineligible for autologous stem cell transplantation. Bortezomib has also been incorporated into conditioning regimens before autologous stem cell transplantation, as well as into post-ASCT consolidation therapy, and in the maintenance setting. In addition, a new route of bortezomib administration, subcutaneous infusion, has recently been approved. Recently, several new agents have been introduced into the clinic, including carfilzomib, marizomib, and MLN9708, and trials investigating these "second-generation" PIs in patients with relapsed/refractory MMs have demonstrated positive results. This review provides an overview of the role of PIs in the treatment of MM, focusing on developments over the past decade.


Subject(s)
Cysteine Proteinase Inhibitors/therapeutic use , Medical Oncology/trends , Multiple Myeloma/drug therapy , Proteasome Inhibitors , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Boronic Acids/adverse effects , Boronic Acids/pharmacology , Boronic Acids/therapeutic use , Bortezomib , Cysteine Proteinase Inhibitors/adverse effects , Cysteine Proteinase Inhibitors/pharmacology , Humans , Medical Oncology/methods , Pyrazines/adverse effects , Pyrazines/pharmacology , Pyrazines/therapeutic use , Time Factors
8.
CNS Neurol Disord Drug Targets ; 11(6): 768-73, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22483304

ABSTRACT

Nurr1 is a member of the nuclear receptor superfamily and is a potential susceptibility gene for Parkinson's disease (PD). Several lines of studies in vitro and in vivo reported that defects in the Nurr1 gene cause nigrostriatal neuronal deficiency as seen in PD. In the present study, we used a a synthetic low molecular weight Nurr1 activator which increases the transcription of Nurr1 to investigate whether it has anti-parkinsonian effects against nigrostriatal neuronal degeneration induced by proteasome inhibitor lactacystin. Adult C57BL/6 mice were treated orally with the Nurr1 activator and an inactive structural analog as a control at a dose of 10mg/kg per day, starting 3 days before microinjection of proteasome inhibitor lactacystin into the medial forebrain bundle and the treatment continued for a total of 4 weeks. Animal behavior tests, and pathological and biochemical examinations were performed to determine the anti-parkinsonian effects of the Nurr1 activator. We found that treatment with the Nurr1 activator significantly improved rotarod performance, attenuated dopamine neuron loss and nigrostriatal dopamine reduction, increased expression of Nurr1, dopamine transporter and vesicular monoamine transporter 2, and alleviated microglial activation in the substantia nigra of lactacystin-lesioned mice. These results suggest that the Nurr1 activator may become an innovative strategy for the treatment of PD.


Subject(s)
Dopamine/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Parkinsonian Disorders/drug therapy , Acetylcysteine/adverse effects , Acetylcysteine/analogs & derivatives , Animals , Cysteine Proteinase Inhibitors/adverse effects , Disease Models, Animal , Dopamine Plasma Membrane Transport Proteins/drug effects , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopaminergic Neurons/drug effects , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Motor Activity/drug effects , Neostriatum/drug effects , Neostriatum/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/drug effects , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Parkinsonian Disorders/chemically induced , Proteasome Endopeptidase Complex/drug effects , RNA, Messenger/analysis , Real-Time Polymerase Chain Reaction , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Transcriptional Activation , Vesicular Monoamine Transport Proteins/drug effects , Vesicular Monoamine Transport Proteins/metabolism
9.
Clin Exp Immunol ; 168(3): 268-73, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22519588

ABSTRACT

Major histocompatibility complex (MHC) class I-restricted T cell epitopes are generated mainly by the immunoproteasome in antigen-presenting cells. Therefore, inhibition of activity of this proteolytic complex molecule is thought to be a potential treatment for cell-mediated autoimmune diseases. We therefore studied the efficacy of an immunoproteasome inhibitor, ONX 0914 (formerly PR-957), for the treatment of autoimmune thyroid diseases, including cell-mediated Hashimoto's thyroiditis and autoantibody-mediated Graves' hyperthyroidism using mouse models. Our data show that ONX 0914 was effective prophylactically and therapeutically at suppressing the degree of intrathyroidal lymphocyte infiltration and, to a lesser degree, the titres of anti-thyroglobulin autoantibodies in non-obese diabetic (NOD)-H2(h4) mice, an iodine-induced autoimmune thyroiditis model. It also inhibited differentiation of T cells to T helper type 1 (Th1) and Th17 cells, effector T cell subsets critical for development of thyroiditis in this mouse strain. In contrast, its effect on the Graves' model was negligible. Although ONX 0914 exerts its immune-suppressive effect through not only suppression of immune proteasome but also other mechanism(s), such as inhibition of T cell differentiation, the present results suggest that the immunoproteasome is a novel drug target in treatment of Hashimoto's thyroiditis in particular and cell-mediated autoimmune diseases in general.


Subject(s)
Cysteine Proteinase Inhibitors/therapeutic use , Graves Disease/drug therapy , Hashimoto Disease/drug therapy , Oligopeptides/therapeutic use , Proteasome Inhibitors , Th1 Cells/drug effects , Th17 Cells/drug effects , Animals , Antibody Formation/drug effects , Autoantibodies/blood , Cells, Cultured , Cysteine Proteinase Inhibitors/administration & dosage , Cysteine Proteinase Inhibitors/adverse effects , Disease Models, Animal , Graves Disease/immunology , Hashimoto Disease/chemically induced , Hashimoto Disease/immunology , Humans , Immunity, Cellular/drug effects , Iodine/administration & dosage , Mice , Mice, Inbred NOD , Oligopeptides/administration & dosage , Oligopeptides/adverse effects , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/pathology , Th17 Cells/immunology , Th17 Cells/metabolism , Th17 Cells/pathology , Thyroglobulin/immunology , Thyroid Gland/drug effects , Thyroid Gland/pathology
11.
Free Radic Biol Med ; 49(8): 1290-7, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20678570

ABSTRACT

Maintaining protein homeostasis is vital to cell viability, with numerous studies demonstrating a role for proteasome inhibition occurring during the aging of a variety of tissues and, presumably, contributing to the disruption of cellular homeostasis during aging. In this study we sought to elucidate the differences between neurons and astrocytes in regard to basal levels of protein synthesis, proteasome-mediated protein degradation, and sensitivity to cytotoxicity after proteasome inhibitor treatment. In these studies we demonstrate that neurons have an increased vulnerability, compared to astrocyte cultures, to proteasome-inhibitor-induced cytotoxicity. No significant difference was observed between these two cell types in regard to the basal rates of protein synthesis, or basal rates of protein degradation, in the pool of short-lived proteins. After proteasome inhibitor treatment neuronal crude lysates were observed to undergo greater increases in the levels of ubiquitinated and oxidized proteins and selectively exhibited increased levels of newly synthesized proteins accumulating within the insoluble protein pool, compared to astrocytes. Together, these data suggest a role for increased oxidized proteins and sequestration of newly synthesized proteins in the insoluble protein pool, as potential mediators of the selective neurotoxicity after proteasome inhibitor treatment. The implications for neurons exhibiting increased sensitivity to acute proteasome inhibitor exposure, and the corresponding changes in protein homeostasis observed after proteasome inhibition, are discussed in the context of both aging and age-related disorders of the nervous system.


Subject(s)
Astrocytes/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Leupeptins/pharmacology , Neurons/metabolism , Oxidative Stress , Aging/drug effects , Aging/metabolism , Aging/pathology , Animals , Astrocytes/drug effects , Astrocytes/pathology , Cell Extracts , Cells, Cultured , Cysteine Proteinase Inhibitors/adverse effects , Cysteine Proteinase Inhibitors/therapeutic use , Humans , Leupeptins/adverse effects , Leupeptins/therapeutic use , Neurons/drug effects , Neurons/pathology , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/prevention & control , Oxidation-Reduction/drug effects , Proteasome Inhibitors , Rats , Rats, Sprague-Dawley , Solubility/drug effects , Ubiquitination/drug effects
12.
Mol Ther ; 18(6): 1143-54, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20372109

ABSTRACT

Bacterial superantigen (BSAg)-induced toxic shock syndrome (TSS) and bacterial lipopolysaccharide (LPS)-induced shock are characterized by severe systemic inflammation. As nuclear factor kappaB (NF kappaB) plays an important role in inflammation and bortezomib, a proteasome inhibitor widely used in cancer chemotherapy, is a potent inhibitor of NF kappaB activation, we evaluated the therapeutic and prophylactic use of bortezomib in these conditions using murine models. Bortezomib prophylaxis significantly reduced serum levels of many cytokines and chemokines induced by BSAg. However, at 3 hours, serum level of TNF-a, an important cytokine implicated in TSS, was significantly reduced but not abolished. At 6 hours, there was no difference in the serum TNF-a levels between bortezomib treated and untreated mice challenged with staphylococcal enterotoxin B (SEB). Paradoxically, all mice treated with bortezomib either before or after BSAg challenge succumbed to TSS. Neither bortezomib nor BSAg was lethal if given alone. Serum biochemical parameters and histopathological findings suggested acute liver failure as the possible cause of mortality. Liver tissue from SEB-challenged mice treated with bortezomib showed a significant reduction in NF kappaB activation. Because NF kappaB-dependent antiapoptotic pathways protect hepatocytes from TNF-alpha-induced cell death, inhibition of NF kappaB brought forth by bortezomib in the face of elevated TNF-alpha levels caused by BSAg or LPS is detrimental.


Subject(s)
Boronic Acids/adverse effects , Cysteine Proteinase Inhibitors/adverse effects , Lipopolysaccharides/toxicity , Proteasome Inhibitors , Pyrazines/adverse effects , Superantigens/toxicity , Systemic Inflammatory Response Syndrome/chemically induced , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Animals , Apoptosis , Bortezomib , Cytokines/biosynthesis , HLA-DR3 Antigen/genetics , Liver/drug effects , Liver/physiopathology , Mice , Mice, Transgenic , NF-kappa B/metabolism
13.
Ross Fiziol Zh Im I M Sechenova ; 96(12): 1190-202, 2010 Dec.
Article in Russian | MEDLINE | ID: mdl-21473106

ABSTRACT

A decrease in activity of ubiquitin proteasome system results in accumulation of toxic forms of protein and cell degeneration, including dopamine (DA)-ergic neurons in the substantia nigra; these neurons are remarkable for their low proteolytic activity of proteosomes that makes them more vulnerable, especially when subjected to the neurotoxin action or Parkinson's disease (PD). The goal of the present study is to develop a model on the basis of inhibition of proteasome activity of nigral cell degeneration which is not accompanied by disturbances in motor behavior but leads to changes in sleep-wake cycle characteristic of the non-motor behaviour. We determined the optimal dose of natural inhibitor of proteasome lactacystin (0.4 mkg) and developed a preclinical model of PD in Wistar rats. We established that on the 14th day following lactacystin double (with one-week interval) bilateral injection into the substantia nigra the developing effects involved 28 % degeneration of DA-ergic neurons in the compact part of the substantia nigra, absence of disorders in motor behaviour, and increase in the total time of rapid eye movement sleep by 37 % at the second half of inactive day phase. These data and an increase in the level of key enzyme of DA synthesis tyrosine hydroxylase (TH) in survived neurons in the substantia nigra as well as the presence of the inverse correlation dependency (r = -0.8, p < 0.01) between the number of survived neurons and the level of TH inside them suggest a hypothesis that the increase in the duration of rapid eye movement sleep could be a non-motor marker of the preclinical stage of PD reflecting a reservation of compensatory potentials in the nigrostriatal system.


Subject(s)
Acetylcysteine/analogs & derivatives , Cysteine Proteinase Inhibitors/adverse effects , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Proteasome Inhibitors , Sleep Stages/drug effects , Substantia Nigra/metabolism , Acetylcysteine/adverse effects , Acetylcysteine/pharmacology , Animals , Cysteine Proteinase Inhibitors/pharmacology , Dopamine/metabolism , Male , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Parkinson Disease, Secondary/physiopathology , Rats , Rats, Wistar , Substantia Nigra/pathology , Substantia Nigra/physiopathology , Tyrosine 3-Monooxygenase/metabolism
14.
Curr Opin Drug Discov Devel ; 11(5): 616-25, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18729013

ABSTRACT

The proteasome is a multicatalytic protease complex that mediates the controlled degradation of intracellular proteins, including key components of pathways that contribute to cancer cell growth and immune cell signaling. Validation for the proteasome as a therapeutic target in oncology was provided by bortezomib, a proteasome inhibitor that was approved for the treatment of multiple myeloma in 2003. Since that time, a number of structurally and mechanistically distinct proteasome inhibitors have entered clinical development in oncology. In this review, the chemical properties, preclinical antitumor activities and early clinical trials of these next-generation proteasome inhibitors are described and the potential for future proteasome inhibitor development in autoimmune indications is discussed.


Subject(s)
Antineoplastic Agents/therapeutic use , Cysteine Proteinase Inhibitors/therapeutic use , Drug Design , Immunologic Factors/therapeutic use , Proteasome Inhibitors , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemistry , Autoimmune Diseases/drug therapy , Cysteine Proteinase Inhibitors/adverse effects , Cysteine Proteinase Inhibitors/chemistry , Drug Evaluation, Preclinical , Humans , Immunologic Factors/adverse effects , Immunologic Factors/chemistry , Molecular Structure , Neoplasms/drug therapy , Proteasome Endopeptidase Complex/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship , Treatment Outcome
15.
Exp Biol Med (Maywood) ; 233(7): 881-90, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18445767

ABSTRACT

The impairment of ubiquitin-proteasome system (UPS) is a cellular mechanism underlying the neurodegenerative process in Parkinson's disease (PD). Glial cell line-derived neurotrophic factor (GDNF) is one of the most potent neurotrophic factors promoting the growth and survival of mesencephalic dopamine (DA) neurons. To investigate whether GDNF has neuroprotective effects in a PD model induced by UPS impairment we administered GDNF by osmotic pump in C57BL/6 mice after nigrostriatal lesions with stereotactic injection of proteasome inhibitor lactacystin in the middle forebrain bundle. We found that lactacystin injection severely injured the nigral DA neurons and reduced the striatal levels of DA and its metabolites, while prolonged administration of GDNF at a sustained moderate dose for two weeks can significantly attenuate the lactacystin-induced loss of nigral DA neurons and striatal DA levels by 31% and 40%, respectively. We also investigated the molecular mechanisms for the neuroprotective effects of GDNF showing that lactacystin administration can cause the phosphorylation of extracellular signal-regulated kinase (ERK), p38MAPK (p38), and the c-Jun N-terminal kinase (JNK), whereas GDNF treatment can further enhance the phosphorylation of ERK and Akt but reduce the levels of JNK and p38. These results indicate that prolonged treatment with GDNF can protect the nigral DA neurons from the UPS impairment-induced degeneration. Several signaling path-ways including p38, JNK, Akt and ERK molecules seem to play an important role in this neuroprotection by GDNF.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/therapeutic use , Nerve Degeneration/prevention & control , Parkinson Disease/prevention & control , Proteasome Inhibitors , Signal Transduction/physiology , Substantia Nigra/pathology , Acetylcysteine/adverse effects , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Animals , Cysteine Proteinase Inhibitors/adverse effects , Cysteine Proteinase Inhibitors/pharmacology , Disease Models, Animal , Dopamine/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase Kinases/metabolism , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Parkinson Disease/etiology , Parkinson Disease/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Random Allocation , Signal Transduction/drug effects , Substantia Nigra/drug effects , Substantia Nigra/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Neurology ; 63(3): 547-9, 2004 Aug 10.
Article in English | MEDLINE | ID: mdl-15304592

ABSTRACT

Minocycline is an antibiotic with anti-inflammatory and antiapoptotic properties that prolongs survival in a transgenic Huntington disease (HD) mouse model. In a double-blind, randomized, placebo-controlled study of minocycline in 60 HD patients, the authors determined that over 8 weeks, minocycline at 100 and 200 mg/day was well tolerated and safe in HD patients. Tolerability and adverse event frequency were similar between treatment and placebo groups.


Subject(s)
Cysteine Proteinase Inhibitors/therapeutic use , Huntington Disease/drug therapy , Minocycline/therapeutic use , Adult , Apoptosis/drug effects , Caspase 3 , Caspase Inhibitors , Cysteine Proteinase Inhibitors/administration & dosage , Cysteine Proteinase Inhibitors/adverse effects , Dose-Response Relationship, Drug , Double-Blind Method , Female , Gastrointestinal Diseases/chemically induced , Humans , Male , Middle Aged , Minocycline/administration & dosage , Minocycline/adverse effects , Patient Dropouts , Treatment Failure , Treatment Outcome
17.
Crit Care Med ; 30(8): 1722-8, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12163783

ABSTRACT

OBJECTIVE: To investigate the efficacy and the safety of the parenteral administration of C1-inhibitor to patients with severe sepsis or septic shock. DESIGN: Double blind, randomized, and placebo-controlled trial. SETTING: Surgical and medical intensive care units of a tertiary care university hospital. PATIENTS: Forty consecutive patients (20 C1-inhibitor/20 placebo) who entered the intensive care unit with severe sepsis or septic shock. INTERVENTION: C1-inhibitor intravenously in a 1-hr infusion, starting with 6000 IU, followed by 3000 IU, 2000 IU, and 1000 IU at 12-hr intervals, compared with placebo. MEASUREMENTS AND MAIN RESULTS: C1-inhibitor administration significantly increased plasma C1-inhibitor antigen and activity levels during days 1-4 (p <.007). Patients in the C1-inhibitor group had significantly lower serum creatinine concentrations on day 3 (p =.048) and 4 (p =.01) than placebo patients. Multiple organ dysfunction assessed by logistic organ dysfunction and sepsis-related organ failure assessment scores was less pronounced in patients treated with C1-inhibitor. Mortality rate was similar in both groups. There were no C1-inhibitor-related side effects. CONCLUSIONS: C1-inhibitor administration attenuated renal impairment in patients with severe sepsis or septic shock.


Subject(s)
Complement C1 Inactivator Proteins/administration & dosage , Cysteine Proteinase Inhibitors/administration & dosage , Shock, Septic/drug therapy , Staphylococcal Infections/drug therapy , Streptococcal Infections/drug therapy , Adult , Aged , Blood Urea Nitrogen , Complement C1 Inactivator Proteins/adverse effects , Complement C1 Inhibitor Protein , Coronary Artery Disease/complications , Coronary Artery Disease/epidemiology , Creatinine/blood , Cysteine Proteinase Inhibitors/adverse effects , Double-Blind Method , Female , Humans , Infusions, Parenteral , Kidney/blood supply , Kidney/drug effects , Male , Middle Aged , Prevalence , Renal Dialysis , Renal Insufficiency/etiology , Renal Insufficiency/therapy , Severity of Illness Index , Shock, Septic/complications , Shock, Septic/mortality , Staphylococcal Infections/complications , Staphylococcal Infections/mortality , Statistics as Topic , Streptococcal Infections/complications , Streptococcal Infections/mortality , Survival Analysis , Switzerland/epidemiology , Time Factors , Treatment Outcome
18.
J Neurosci Res ; 69(3): 373-81, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12125078

ABSTRACT

We investigated the effect of two proteasome inhibitors, lactacystin and epoxomicin, on cultured spinal cord neurons. The incubation of spinal neurons with proteasome inhibitors for 24 hr induced neurotoxicity in a dose-dependent manner. We found motor neurons to be more vulnerable to proteasome-induced neurotoxicity than nonmotor neurons. The staining of cell bodies in treated motor neurons was markedly disrupted and showed characteristic granular patterns. Proteasome-induced neurotoxicity is accompanied by apoptotic nuclear changes, posttranslational modification of the cellular proteins, generation of intracellular free radicals, reduction in the amount of reduced glutathione, and mitochondrial dysfunction. Neurotoxicity was reduced by the administration of low concentrations (1-100 nM) of geranylgeranylacetone (GGA), which is widely used as an antiulcer drug, although higher concentrations of this drug produced neurotoxicity in spinal cord neurons. GGA was found to induce the expression of heat shock protein 70 as well as thioredoxin, which may partly contribute to the protective effect of GGA. These data suggest that the inhibition of proteasome may play a role in the mechanism of neurodegenerative diseases of the spinal cord, such as amyotrophic lateral sclerosis, and that the use of GGA may be effective in the treatment of these conditions.


Subject(s)
Acetylcysteine/analogs & derivatives , Cysteine Endopeptidases/metabolism , Diterpenes/pharmacology , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/metabolism , Neurons/metabolism , Neuroprotective Agents/pharmacology , Spinal Cord/metabolism , Acetylcysteine/adverse effects , Animals , Cell Survival/drug effects , Cells, Cultured , Cysteine Proteinase Inhibitors/adverse effects , Dose-Response Relationship, Drug , Glutathione/drug effects , Glutathione/metabolism , HSP70 Heat-Shock Proteins/metabolism , Immunoblotting , Immunohistochemistry , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/drug effects , Oligopeptides/adverse effects , Proteasome Endopeptidase Complex , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Spinal Cord/cytology , Spinal Cord/drug effects , Thioredoxins/metabolism , Time Factors
19.
J Neurosci Res ; 58(6): 823-30, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10583913

ABSTRACT

Increased levels of reactive oxygen species occur in neurodegenerative disorders and may promote neuron death. The lipid peroxidation product 4-hydroxynonenal (HNE) is increased in neurons following oxidative stress and promotes neuron death in vitro and in vivo. The present study examined the possibility that HNE can increase neuron vulnerability to oxidative stress. Application of low concentrations of HNE (50-500 nM) increased neuron death induced by beta-amyloid or glutamate when added within 3 hr of injury. In addition, treatment with HNE exacerbated mitochondrial reactive oxygen species formation and loss of mitochondrial membrane potential in response to beta-amyloid and glutamate. The ability to exacerbate oxidative stress, mitochondrial dysfunction, and neuron death appears to be specific to HNE, because application of other lipid peroxidation products had no effect. These data indicate a role for low levels of HNE in promoting reactive oxygen species accumulation and neuron degeneration by altering mitochondrial homeostasis. In addition, the present study indicates a possible mechanism for reactive oxygen species and lipid peroxidation toxicity in neurodegenerative conditions.


Subject(s)
Aldehydes/adverse effects , Aldehydes/metabolism , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Amyloid beta-Peptides/adverse effects , Animals , Cell Culture Techniques , Cell Death/drug effects , Cell Death/physiology , Cysteine Proteinase Inhibitors/adverse effects , Cysteine Proteinase Inhibitors/metabolism , Glutamic Acid/adverse effects , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Neurons/cytology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...