Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Pharmacol Rev ; 74(3): 600-629, 2022 07.
Article in English | MEDLINE | ID: mdl-35710131

ABSTRACT

Cathepsin B (CTSB) is a powerful lysosomal protease. This review evaluated CTSB gene knockout (KO) outcomes for amelioration of brain dysfunctions in neurologic diseases and aging animal models. Deletion of the CTSB gene resulted in significant improvements in behavioral deficits, neuropathology, and/or biomarkers in traumatic brain injury, ischemia, inflammatory pain, opiate tolerance, epilepsy, aging, transgenic Alzheimer's disease (AD), and periodontitis AD models as shown in 12 studies. One study found beneficial effects for double CTSB and cathepsin S KO mice in a multiple sclerosis model. Transgenic AD models using amyloid precursor protein (APP) mimicking common sporadic AD in three studies showed that CTSB KO improved memory, neuropathology, and biomarkers; two studies used APP representing rare familial AD and found no CTSB KO effect, and two studies used highly engineered APP constructs and reported slight increases in a biomarker. In clinical studies, all reports found that CTSB enzyme was upregulated in diverse neurologic disorders, including AD in which elevated CTSB was positively correlated with cognitive dysfunction. In a wide range of neurologic animal models, CTSB was also upregulated and not downregulated. Further, human genetic mutation data provided precedence for CTSB upregulation causing disease. Thus, the consilience of data is that CTSB gene KO results in improved brain dysfunction and reduced pathology through blockade of CTSB enzyme upregulation that causes human neurologic disease phenotypes. The overall findings provide strong support for CTSB as a rational drug target and for CTSB inhibitors as therapeutic candidates for a wide range of neurologic disorders. SIGNIFICANCE STATEMENT: This review provides a comprehensive compilation of the extensive data on the effects of deleting the cathepsin B (CTSB) gene in neurological and aging mouse models of brain disorders. Mice lacking the CTSB gene display improved neurobehavioral deficits, reduced neuropathology, and amelioration of neuronal cell death and inflammatory biomarkers. The significance of the compelling CTSB evidence is that the data consilience validates CTSB as a drug target for discovery of CTSB inhibitors as potential therapeutics for treating numerous neurological diseases.


Subject(s)
Alzheimer Disease , Cathepsin B , Alzheimer Disease/metabolism , Animals , Cathepsin B/genetics , Cathepsin B/metabolism , Disease Models, Animal , Gene Knockout Techniques , Humans , Mice , Mice, Knockout , Mice, Transgenic
2.
Biochemistry ; 61(4): 228-238, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35119840

ABSTRACT

CA-074 is a selective inhibitor of cathepsin B, a lysosomal cysteine protease. CA-074 has been utilized in numerous studies to demonstrate the role of this protease in cellular and physiological functions. Cathepsin B in numerous human disease mechanisms involves its translocation from acidic lysosomes of pH 4.6 to neutral pH 7.2 of cellular locations, including the cytosol and extracellular environment. To gain in-depth knowledge of CA-074 inhibition under these different pH conditions, this study evaluated the molecular features, potency, and selectivity of CA-074 for cathepsin B inhibition under acidic and neutral pH conditions. This study demonstrated that CA-074 is most effective at inhibiting cathepsin B at an acidic pH of 4.6 with nM potency, which was more than 100-fold more potent than its inhibition at a neutral pH of 7.2. The pH-dependent inhibition of CA-074 was abolished by methylation of its C-terminal proline, indicating the requirement for the free C-terminal carboxyl group for pH-dependent inhibition. Under these acidic and neutral pH conditions, CA-074 maintained its specificity for cathepsin B over other cysteine cathepsins, displayed irreversible inhibition, and inhibited diverse cleavages of peptide substrates of cathepsin B assessed by profiling mass spectrometry. Molecular docking suggested that pH-dependent ionic interactions of the C-terminal carboxylate of CA-074 occur with His110 and His111 residues in the S2' subsite of the enzyme at pH 4.6, but these interactions differ at pH 7.2. While high levels of CA-074 or CA-074Me (converted by cellular esterases to CA-074) are used in biological studies to inhibit cathepsin B at both acidic and neutral pH locations, it is possible that adjusted levels of CA-074 or CA-074Me may be explored to differentially affect cathepsin B activity at these different pH values. Overall, the results of this study demonstrate the molecular, kinetic, and protease specificity features of CA-074 pH-dependent inhibition of cathepsin B.


Subject(s)
Cathepsin B/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Animals , Cathepsin B/metabolism , Cathepsin L/pharmacology , Cathepsins/metabolism , Cysteine/metabolism , Cysteine Proteinase Inhibitors/chemistry , Cytosol/metabolism , Dipeptides/chemistry , Humans , Hydrogen-Ion Concentration , Kinetics , Lysosomes/metabolism , Mass Spectrometry/methods , Molecular Docking Simulation , Peptides/metabolism
3.
J Neurochem ; 155(3): 300-312, 2020 11.
Article in English | MEDLINE | ID: mdl-32330298

ABSTRACT

Lysosomes are known to mediate neurite outgrowth in neurons. However, the principal lysosomal molecule controlling that outgrowth is unclear. We studied primary mouse neurons in vitro and found that they naturally develop neurite outgrowths over time and as they did so the lysosomal cysteine protease cathepsin B (CTSB) mRNA levels dramatically increased. Surprisingly, we found that treating those neurons with CA-074Me, which inhibits CTSB, prevented neurites. As that compound also inhibits another protease, we evaluated a N2a neuronal cell line in which the CTSB gene was deleted (CTSB knockout, KO) using CRISPR technology and induced their neurite outgrowth by treatment with retinoic acid. We found that CTSB KO N2a cells failed to produce neurite outgrowths but the wild-type (WT) did. CA-074Me is a cell permeable prodrug of CA-074, which is cell impermeable and a specific CTSB inhibitor. Neurite outgrowth was and was not suppressed in WT N2a cells treated with CA-074Me and CA-074, respectively. Lysosome-associated membrane glycoprotein 2-positive lysosomes traffic to the plasma cell membrane in WT but not in CTSB KO N2 a cells. Interestingly, no obvious differences between WT and CTSB KO N2a cells were found in neurite outgrowth regulatory proteins, PI3K/AKT, ERK/MAPK, cJUN, and CREB. These findings show that intracellular CTSB controls neurite outgrowth and that it does so through regulation of lysosomal trafficking and remodeling in neurons. This adds valuable information regarding the physiological function of CTSB in neural development.


Subject(s)
Cathepsin B/deficiency , Lysosomes/metabolism , Neurites/metabolism , Neurons/metabolism , Animals , Animals, Newborn , Cathepsin B/antagonists & inhibitors , Cathepsin B/genetics , Cell Line, Tumor , Cells, Cultured , Cysteine Proteases/deficiency , Cysteine Proteases/genetics , Female , Gene Knockout Techniques/methods , Male , Mice , Mice, Inbred C57BL , Neocortex/cytology , Neocortex/metabolism , Protein Transport/physiology
4.
Biochim Biophys Acta ; 1824(1): 89-104, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21925292

ABSTRACT

Recent new findings indicate significant biological roles of cysteine cathepsin proteases in secretory vesicles for production of biologically active peptides. Notably, cathepsin L in secretory vesicles functions as a key protease for proteolytic processing of proneuropeptides (and prohormones) into active neuropeptides that are released to mediate cell-cell communication in the nervous system for neurotransmission. Moreover, cathepsin B in secretory vesicles has been recently identified as a ß-secretase for production of neurotoxic ß- amyloid (Aß) peptides that accumulate in Alzheimer's disease (AD), participating as a notable factor in the severe memory loss in AD. These secretory vesicle functions of cathepsins L and B for production of biologically active peptides contrast with the well-known role of cathepsin proteases in lysosomes for the degradation of proteins to result in their inactivation. The unique secretory vesicle proteome indicates proteins of distinct functional categories that provide the intravesicular environment for support of cysteine cathepsin functions. Features of the secretory vesicle protein systems insure optimized intravesicular conditions that support the proteolytic activity of cathepsins. These new findings of recently discovered biological roles of cathepsins L and B indicate their significance in human health and disease. This article is part of a Special Issue entitled: Proteolysis 50 years after the discovery of lysosome.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Cathepsins/physiology , Neurotransmitter Agents/metabolism , Peptides/metabolism , Secretory Vesicles/metabolism , Alzheimer Disease/etiology , Alzheimer Disease/genetics , Amino Acid Sequence , Animals , Cathepsin B/chemistry , Cathepsin B/genetics , Cathepsin B/metabolism , Cathepsin B/physiology , Cathepsin L/chemistry , Cathepsin L/genetics , Cathepsin L/metabolism , Cathepsin L/physiology , Cathepsins/chemistry , Cathepsins/genetics , Cathepsins/metabolism , Cysteine Proteases/chemistry , Cysteine Proteases/genetics , Cysteine Proteases/metabolism , Cysteine Proteases/physiology , Humans , Models, Biological , Molecular Sequence Data , Proteolysis , Secretory Vesicles/enzymology
5.
J Alzheimers Dis ; 93(1): 33-46, 2023.
Article in English | MEDLINE | ID: mdl-36970896

ABSTRACT

The lysosomal cysteine protease cathepsin B (CTSB) has been suggested as a biomarker for Alzheimer's disease (AD) because elevated serum CTSB in AD patients has been found to correlate with cognitive dysfunction. Furthermore, CTSB gene knockout (KO) in non-transgenic and transgenic AD animal models showed that elimination of CTSB improved memory deficits. However, conflicting CTSB KO results on amyloid-ß (Aß) pathology in transgenic AD models have been reported. The conflict is resolved here as likely being due to the different hAßPP transgenes used in the different AD mouse models. CTSB gene KO reduced wild-type (Wt) ß-secretase activity, brain Aß, pyroglutamate-Aß, amyloid plaque, and memory deficits in models that used cDNA transgenes expressing hAßPP isoform 695. But in models that used mutated mini transgenes expressing hAßPP isoforms 751 and 770, CTSB KO had no effect on Wt ß-secretase activity and slightly increased brain Aß. All models expressed the AßPP transgenes in neurons. These conflicting results in Wt ß-secretase activity models can be explained by hAßPP isoform specific cellular expression, proteolysis, and subcellular processing. CTSB KO had no effect on Swedish mutant (Swe) ß-secretase activity in hAßPP695 and hAßPP751/770 models. Different proteolytic sensitivities for hAßPP with Wt versus Swe ß-secretase site sequences may explain the different CTSB ß-secretase effects in hAßPP695 models. But since the vast majority of sporadic AD patients have Wt ß-secretase activity, the CTSB effects on Swe ß-secretase activity are of little importance to the general AD population. As neurons naturally produce and process hAßPP isoform 695 and not the 751 and 770 isoforms, only the hAßPP695 Wt models mimic the natural neuronal hAßPP processing and Aß production occurring in most AD patients. Significantly, these CTSB KO findings in the hAßPP695 Wt models demonstrate that CTSB participates in memory deficits and production of pyroglutamate-Aß (pyroglu-Aß), which provide rationale for future investigation of CTSB inhibitors in AD therapeutics development.


Subject(s)
Alzheimer Disease , Mice , Animals , Alzheimer Disease/pathology , Cathepsin B/metabolism , Amyloid Precursor Protein Secretases/metabolism , Pyrrolidonecarboxylic Acid , Amyloid beta-Peptides/metabolism , Memory Disorders/genetics , Disease Models, Animal , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Mice, Transgenic
6.
ACS Chem Biol ; 16(9): 1628-1643, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34416110

ABSTRACT

Cathepsin B is a cysteine protease that normally functions within acidic lysosomes for protein degradation, but in numerous human diseases, cathepsin B translocates to the cytosol having neutral pH where the enzyme activates inflammation and cell death. Cathepsin B is active at both the neutral pH 7.2 of the cytosol and the acidic pH 4.6 within lysosomes. We evaluated the hypothesis that cathepsin B may possess pH-dependent cleavage preferences that can be utilized for design of a selective neutral pH inhibitor by (1) analysis of differential cathepsin B cleavage profiles at neutral pH compared to acidic pH using multiplex substrate profiling by mass spectrometry (MSP-MS), (2) design of pH-selective peptide-7-amino-4-methylcoumarin (AMC) substrates, and (3) design and validation of Z-Arg-Lys-acyloxymethyl ketone (AOMK) as a selective neutral pH inhibitor. Cathepsin B displayed preferences for cleaving peptides with Arg in the P2 position at pH 7.2 and Glu in the P2 position at pH 4.6, represented by its primary dipeptidyl carboxypeptidase and modest endopeptidase activity. These properties led to design of the substrate Z-Arg-Lys-AMC having neutral pH selectivity, and its modification with the AOMK warhead to result in the inhibitor Z-Arg-Lys-AOMK. This irreversible inhibitor displays nanomolar potency with 100-fold selectivity for inhibition of cathepsin B at pH 7.2 compared to pH 4.6, shows specificity for cathepsin B over other cysteine cathepsins, and is cell permeable and inhibits intracellular cathepsin B. These findings demonstrate that cathepsin B possesses pH-dependent cleavage properties that can lead to development of a potent, neutral pH inhibitor of this enzyme.


Subject(s)
Cathepsin B/antagonists & inhibitors , Cysteine Proteinase Inhibitors/chemistry , Cytosol/metabolism , Lysosomes/metabolism , Peptides/chemistry , Amino Acid Sequence , Binding Sites , Cathepsins/metabolism , Cell Membrane Permeability , Cysteine Proteinase Inhibitors/metabolism , Endopeptidases/metabolism , Humans , Hydrogen-Ion Concentration , Kinetics , Mass Spectrometry , Peptides/metabolism , Protein Binding , Substrate Specificity
7.
Biol Chem ; 391(8): 861-72, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20536395

ABSTRACT

Beta-amyloid (Abeta) in the brain is a major factor involved in Alzheimer's disease (AD) that results in severe memory deficit. Our recent studies demonstrate pharmacogenetic differences in the effects of inhibitors of cathepsin B to improve memory and reduce Abeta in different mouse models of AD. The inhibitors improve memory and reduce brain Abeta in mice expressing the wild-type (WT) beta-secretase site of human APP, expressed in most AD patients. However, these inhibitors have no effect in mice expressing the rare Swedish (Swe) mutant amyloid precursor protein (APP). Knockout of the cathepsin B decreased brain Abeta in mice expressing WT APP, validating cathepsin B as the target. The specificity of cathepsin B to cleave the WT beta-secretase site, but not the Swe mutant site, of APP for Abeta production explains the distinct inhibitor responses in the different AD mouse models. In contrast to cathepsin B, the BACE1 beta-secretase prefers to cleave the Swe mutant site. Discussion of BACE1 data in the field indicate that they do not preclude cathepsin B as also being a beta-secretase. Cathepsin B and BACE1 could participate jointly as beta-secretases. Significantly, the majority of AD patients express WT APP and, therefore, inhibitors of cathepsin B represent candidate drugs for AD.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Cathepsin B/antagonists & inhibitors , Drug Design , Protease Inhibitors/pharmacokinetics , Protease Inhibitors/therapeutic use , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Cathepsin B/genetics , Disease Models, Animal , Humans , Mice , Mice, Knockout , Neurons/metabolism , Protease Inhibitors/pharmacology , Secretory Pathway/drug effects , Substrate Specificity
8.
Biochim Biophys Acta Proteins Proteom ; 1868(8): 140428, 2020 08.
Article in English | MEDLINE | ID: mdl-32305689

ABSTRACT

Investigations of Alzheimer's disease (AD), traumatic brain injury (TBI), and related brain disorders have provided extensive evidence for involvement of cathepsin B, a lysosomal cysteine protease, in mediating the behavioral deficits and neuropathology of these neurodegenerative diseases. This review integrates findings of cathepsin B regulation in clinical biomarker studies, animal model genetic and inhibitor evaluations, structural studies, and lysosomal cell biological mechanisms in AD, TBI, and related brain disorders. The results together indicate the role of cathepsin B in the behavioral deficits and neuropathology of these disorders. Lysosomal leakage occurs in AD and TBI, and related neurodegeneration, which leads to the hypothesis that cathepsin B is redistributed from the lysosome to the cytosol where it initiates cell death and inflammation processes associated with neurodegeneration. These results together implicate cathepsin B as a major contributor to these neuropathological changes and behavioral deficits. These findings support the investigation of cathepsin B as a potential drug target for therapeutic discovery and treatment of AD, TBI, and TBI-related brain disorders.


Subject(s)
Alzheimer Disease/enzymology , Brain Injuries, Traumatic/enzymology , Brain/enzymology , Cathepsin B/genetics , Neurocognitive Disorders/enzymology , Neurons/enzymology , Adult , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Brain/drug effects , Brain/pathology , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/pathology , Cathepsin B/antagonists & inhibitors , Cathepsin B/metabolism , Cell Death/drug effects , Cell Death/genetics , Child , Cytosol/drug effects , Cytosol/enzymology , Disease Models, Animal , Fetus , Gene Expression Regulation , Humans , Infant , Lysosomes/drug effects , Lysosomes/enzymology , Molecular Targeted Therapy , Neurocognitive Disorders/drug therapy , Neurocognitive Disorders/genetics , Neurocognitive Disorders/pathology , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/therapeutic use , Signal Transduction
9.
J Neurotrauma ; 37(13): 1574-1586, 2020 07 01.
Article in English | MEDLINE | ID: mdl-31973644

ABSTRACT

Cathepsin B (CatB), a lysosomal cysteine protease, is important to brain function and may have dual utility as a peripheral biomarker of moderate-severe traumatic brain injury (TBI). The present study determined levels of pro- and mature (mat) CatB protein as well as cysteine protease activity within the frontal cortex (FC; proximal injury site), hippocampus (HC; distal injury site), and cerebral spinal fluid (CSF) collected 1-7 days after craniotomy and penetrating ballistic-like brain injury (PBBI) in rats. Values were compared with naïve controls. Further, the utility of CatB protein as a translational biomarker was determined in CSF derived from patients with severe TBI. Craniotomy increased matCatB levels in the FC and HC, and led to elevation of HC activity at day 7. PBBI caused an even greater elevation in matCatB within the FC and HC within 3-7 days. After PBBI, cysteine protease activity peaked at 3 days in the FC and was elevated at 1 day and 7 days, but not 3 days, in the HC. In rat CSF, proCatB, matCatB, and cysteine protease activity peaked at 3 days after craniotomy and PBBI. Addition of CA-074, a CatB-specific inhibitor, confirmed that protease activity was due to active matCatB in rat brain tissues and CSF at all time-points. In patients, CatB protein was detectable from 6 h through 10 days after TBI. Notably, CatB levels were significantly higher in CSF collected within 3 days after TBI compared with non-TBI controls. Collectively, this work indicates that CatB and its cysteine protease activity may serve as collective molecular signatures of TBI progression that differentially vary within both proximal and distal brain regions. CatB and its protease activity may have utility as a surrogate, translational biomarker of acute-subacute TBI.


Subject(s)
Brain Injuries, Traumatic/metabolism , Brain/metabolism , Cathepsin B/metabolism , Cysteine Proteases/metabolism , Head Injuries, Penetrating/metabolism , Animals , Biomarkers/cerebrospinal fluid , Biomarkers/metabolism , Brain Injuries, Traumatic/cerebrospinal fluid , Cathepsin B/cerebrospinal fluid , Craniotomy/adverse effects , Cysteine Proteases/cerebrospinal fluid , Enzyme Activation/physiology , Head Injuries, Penetrating/cerebrospinal fluid , Humans , Male , Rats , Rats, Sprague-Dawley
10.
Biochem Biophys Res Commun ; 386(2): 284-8, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19501042

ABSTRACT

Neurotoxic beta-amyloid (Abeta) peptides participate in Alzheimer's disease (AD); therefore, reduction of Abeta generated from APP may provide a therapeutic approach for AD. Gene knockout studies in transgenic mice producing human Abeta may identify targets for reducing Abeta. This study shows that knockout of the cathepsin B gene in mice expressing human wild-type APP (hAPPwt) results in substantial decreases in brain Abeta40 and Abeta42 by 67% and decreases in levels of the C-terminal beta-secretase fragment (CTFbeta) derived from APP. In contrast, knockout of cathepsin B in mice expressing hAPP with the rare Swedish (Swe) and Indiana (Ind) mutations had no effect on Abeta. The difference in reduction of Abeta in hAPPwt mice, but not in hAPPSwe/Ind mice, shows that the transgenic model can affect cathepsin B gene knockout results. Since most AD patients express hAPPwt, these data validate cathepsin B as a target for development of inhibitors to lower Abeta in AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Cathepsin B/deficiency , Peptide Fragments/metabolism , Alzheimer Disease/therapy , Amyloid beta-Protein Precursor/genetics , Animals , Cathepsin B/genetics , Gene Knockout Techniques , Humans , Mice , Mice, Transgenic , Mutation
11.
Front Neurol ; 6: 178, 2015.
Article in English | MEDLINE | ID: mdl-26388830

ABSTRACT

There is currently no therapeutic drug treatment for traumatic brain injury (TBI) despite decades of experimental clinical trials. This may be because the mechanistic pathways for improving TBI outcomes have yet to be identified and exploited. As such, there remains a need to seek out new molecular targets and their drug candidates to find new treatments for TBI. This review presents supporting evidence for cathepsin B, a cysteine protease, as a potentially important drug target for TBI. Cathepsin B expression is greatly up-regulated in TBI animal models, as well as in trauma patients. Importantly, knockout of the cathepsin B gene in TBI mice results in substantial improvements of TBI-caused deficits in behavior, pathology, and biomarkers, as well as improvements in related injury models. During the process of TBI-induced injury, cathepsin B likely escapes the lysosome, its normal subcellular location, into the cytoplasm or extracellular matrix (ECM) where the unleashed proteolytic power causes destruction via necrotic, apoptotic, autophagic, and activated glia-induced cell death, together with ECM breakdown and inflammation. Significantly, chemical inhibitors of cathepsin B are effective for improving deficits in TBI and related injuries including ischemia, cerebral bleeding, cerebral aneurysm, edema, pain, infection, rheumatoid arthritis, epilepsy, Huntington's disease, multiple sclerosis, and Alzheimer's disease. The inhibitor E64d is unique among cathepsin B inhibitors in being the only compound to have demonstrated oral efficacy in a TBI model and prior safe use in man and as such it is an excellent tool compound for preclinical testing and clinical compound development. These data support the conclusion that drug development of cathepsin B inhibitors for TBI treatment should be accelerated.

12.
J Alzheimers Dis ; 41(1): 129-49, 2014.
Article in English | MEDLINE | ID: mdl-24595198

ABSTRACT

Pyroglutamate amyloid-ß peptides (pGlu-Aß) are particularly pernicious forms of amyloid-ß peptides (Aß) present in Alzheimer's disease (AD) brains. pGlu-Aß peptides are N-terminally truncated forms of full-length Aß peptides (flAß(1-40/42)) in which the N-terminal glutamate is cyclized to pyroglutamate to generate pGlu-Aß(3-40/42). ß-secretase cleavage of amyloid-ß precursor protein (AßPP) produces flAß(1-40/42), but it is not yet known whether the ß-secretase BACE1 or the alternative ß-secretase cathepsin B (CatB) participate in the production of pGlu-Aß. Therefore, this study examined the effects of gene knockout of these proteases on brain pGlu-Aß levels in transgenic AßPPLon mice, which express AßPP isoform 695 and have the wild-type (wt) ß-secretase activity found in most AD patients. Knockout or overexpression of the CatB gene reduced or increased, respectively, pGlu-Aß(3-40/42), flAß(1-40/42), and pGlu-Aß plaque load, but knockout of the BACE1 gene had no effect on those parameters in the transgenic mice. Treatment of AßPPLon mice with E64d, a cysteine protease inhibitor of CatB, also reduced brain pGlu-Aß(3-42), flAß(1-40/42), and pGlu-Aß plaque load. Treatment of neuronal-like chromaffin cells with CA074Me, an inhibitor of CatB, resulted in reduced levels of pGlu-Aß(3-40) released from the activity-dependent, regulated secretory pathway. Moreover, CatB knockout and E64d treatment has been previously shown to improve memory deficits in the AßPPLon mice. These data illustrate the role of CatB in producing pGlu-Aß and flAß that participate as key factors in the development of AD. The advantages of CatB inhibitors, especially E64d and its derivatives, as alternatives to BACE1 inhibitors in treating AD patients are discussed.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Brain/drug effects , Brain/physiopathology , Cathepsin B/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Leucine/analogs & derivatives , Pyrrolidonecarboxylic Acid/analysis , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Brain/pathology , Cathepsin B/antagonists & inhibitors , Cathepsin B/genetics , Cattle , Cells, Cultured , Chromaffin Cells/drug effects , Chromaffin Cells/physiology , Dipeptides/pharmacology , Humans , Leucine/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neuroprotective Agents/pharmacology , Peptide Fragments/metabolism , Plaque, Amyloid/drug therapy , Plaque, Amyloid/pathology , Plaque, Amyloid/physiopathology
13.
J Neurotrauma ; 31(5): 515-29, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24083575

ABSTRACT

There are currently no effective therapeutic agents for traumatic brain injury (TBI), but drug treatments for TBI can be developed by validation of new drug targets and demonstration that compounds directed to such targets are efficacious in TBI animal models using a clinically relevant route of drug administration. The cysteine protease, cathepsin B, has been implicated in mediating TBI, but it has not been validated by gene knockout (KO) studies. Therefore, this investigation evaluated mice with deletion of the cathepsin B gene receiving controlled cortical impact TBI trauma. Results indicated that KO of the cathepsin B gene resulted in amelioration of TBI, shown by significant improvement in motor dysfunction, reduced brain lesion volume, greater neuronal density in brain, and lack of increased proapoptotic Bax levels. Notably, oral administration of the small-molecule cysteine protease inhibitor, E64d, immediately after TBI resulted in recovery of TBI-mediated motor dysfunction and reduced the increase in cathepsin B activity induced by TBI. E64d outcomes were as effective as cathepsin B gene deletion for improving TBI. E64d treatment was effective even when administered 8 h after injury, indicating a clinically plausible time period for acute therapeutic intervention. These data demonstrate that a cysteine protease inhibitor can be orally efficacious in a TBI animal model when administered at a clinically relevant time point post-trauma, and that E64d-mediated improvement of TBI is primarily the result of inhibition of cathepsin B activity. These results validate cathepsin B as a new TBI therapeutic target.


Subject(s)
Brain Injuries/drug therapy , Cathepsin B/metabolism , Cysteine Proteinase Inhibitors/therapeutic use , Animals , Brain Injuries/metabolism , Cathepsin B/genetics , Cysteine Proteinase Inhibitors/pharmacology , Disease Models, Animal , Mice , Mice, Knockout
14.
J Alzheimers Dis ; 29(4): 827-40, 2012.
Article in English | MEDLINE | ID: mdl-22337825

ABSTRACT

Therapeutic agents that improve the memory loss of Alzheimer's disease (AD) may eventually be developed if drug targets are identified that improve memory deficits in appropriate AD animal models. One such target is ß-secretase which, in most AD patients, cleaves the wild-type (WT) ß-secretase site sequence of the amyloid-ß protein precursor (AßPP) to produce neurotoxic amyloid-ß (Aß). Thus, an animal model representing most AD patients for evaluating ß-secretase effects on memory deficits is one that expresses human AßPP containing the WT ß-secretase site sequence. BACE1 and cathepsin B (CatB) proteases have ß-secretase activity, but gene knockout studies have not yet validated that the absence of these proteases improves memory deficits in such an animal model. This study assessed the effects of deleting these protease genes on memory deficits in the AD mouse model expressing human AßPP containing the WT ß-secretase site sequence and the London γ-secretase site (AßPPWT/Lon mice). Knockout of the CatB gene in the AßPPWT/Lon mice improved memory deficits and altered the pattern of Aß-related biomarkers in a manner consistent with CatB having WT ß-secretase activity. But deletion of the BACE1 gene had no effect on these parameters in the AßPPWT/Lon mice. These data are the first to show that knockout of a putative ß-secretase gene results in improved memory in an AD animal model expressing the WT ß-secretase site sequence of AßPP, present in the majority of AD patients. CatB may be an effective drug target for improving memory deficits in most AD patients.


Subject(s)
Alzheimer Disease/complications , Amyloid beta-Protein Precursor/metabolism , Cathepsin B/deficiency , Memory Disorders/etiology , Memory Disorders/genetics , Mutation/genetics , Age Factors , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/deficiency , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Aspartic Acid Endopeptidases/deficiency , Brain/metabolism , Brain/pathology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/genetics , Humans , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptide Fragments/metabolism , Plaque, Amyloid/pathology , Reaction Time/genetics
15.
J Alzheimers Dis ; 26(2): 387-408, 2011.
Article in English | MEDLINE | ID: mdl-21613740

ABSTRACT

The cysteine protease cathepsin B is a potential drug target for reducing brain amyloid-ß (Aß) and improving memory in Alzheimer's disease (AD), as reduction of cathepsin B in transgenic mice expressing human wild-type amyloid-ß protein precursor (AßPP) results in significantly decreased brain Aß. Cathepsin B cleaves the wild-type ß-secretase site sequence in AßPP to produce Aß, and cathepsin B inhibitors administered to animal models expressing AßPP containing the wild-type ß-secretase site sequence reduce brain Aß in a manner consistent with ß-secretase inhibition. But such inhibitors could act either by direct inhibition of cathepsin B ß-secretase activity or by off-target inhibition of the other ß-secretase, the aspartyl protease BACE1. To evaluate that issue, we orally administered a cysteine protease inhibitor, E64d, to normal guinea pigs or transgenic mice expressing human AßPP, both of which express the human wild-type ß-secretase site sequence. In guinea pigs, oral E64d administration caused a dose-dependent reduction of up to 92% in brain, CSF, and plasma of Aß40 and Aß42, a reduction of up to 50% in the C-terminal ß-secretase fragment (CTFß), and a 91% reduction in brain cathepsin B activity, but increased brain BACE1 activity by 20%. In transgenic AD mice, oral E64d administration improved memory deficits and reduced brain Aß40 and Aß42, amyloid plaque, brain CTFß, and brain cathepsin B activity, but increased brain BACE1 activity. We conclude that E64d likely reduces brain Aß by inhibiting cathepsin B and not BACE1 ß-secretase activity and that E64d therefore may have potential for treating AD patients.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Aspartic Acid Endopeptidases/metabolism , Brain/drug effects , Cathepsin B/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Leucine/analogs & derivatives , Memory/drug effects , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Brain/metabolism , Brain/pathology , Cathepsin B/metabolism , Cysteine Proteinase Inhibitors/therapeutic use , Disease Models, Animal , Guinea Pigs , Leucine/pharmacology , Leucine/therapeutic use , Mice , Mice, Transgenic
16.
J Biol Chem ; 283(12): 7745-53, 2008 Mar 21.
Article in English | MEDLINE | ID: mdl-18184658

ABSTRACT

Elucidation of Abeta-lowering agents that inhibit processing of the wild-type (WT) beta-secretase amyloid precursor protein (APP) site, present in most Alzheimer disease (AD) patients, is a logical approach for improving memory deficit in AD. The cysteine protease inhibitors CA074Me and E64d were selected by inhibition of beta-secretase activity in regulated secretory vesicles that produce beta-amyloid (Abeta). The regulated secretory vesicle activity, represented by cathepsin B, selectively cleaves the WT beta-secretase site but not the rare Swedish mutant beta-secretase site. In vivo treatment of London APP mice, expressing the WT beta-secretase site, with these inhibitors resulted in substantial improvement in memory deficit assessed by the Morris water maze test. After inhibitor treatment, the improved memory function was accompanied by reduced amyloid plaque load, decreased Abeta40 and Abeta42, and reduced C-terminal beta-secretase fragment derived from APP by beta-secretase. However, the inhibitors had no effects on any of these parameters in mice expressing the Swedish mutant beta-secretase site of APP. The notable efficacy of these inhibitors to improve memory and reduce Abeta in an AD animal model expressing the WT beta-secretase APP site present in the majority of AD patients provides support for CA074Me and E64d inhibitors as potential AD therapeutic agents.


Subject(s)
Alzheimer Disease/enzymology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Protein Precursor/metabolism , Cathepsin B/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Leucine/analogs & derivatives , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Cathepsin B/genetics , Cattle , Cysteine Proteinase Inhibitors/therapeutic use , Dipeptides/therapeutic use , Disease Models, Animal , Drug Evaluation, Preclinical , Gene Expression , Humans , Leucine/pharmacology , Leucine/therapeutic use , Maze Learning/drug effects , Memory/drug effects , Mice , Mice, Transgenic
17.
Biol Chem ; 389(8): 993-1006, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18979625

ABSTRACT

This highlight article describes three Alzheimer's disease (AD) studies presented at the 5th General Meeting of the International Proteolysis Society that address enzymatic mechanisms for producing neurotoxic beta-amyloid (Abeta) peptides. One group described the poor kinetics of BACE 1 for cleaving the wild-type (WT) beta-secretase site of APP found in most AD patients. They showed that cathepsin D displays BACE 1-like specificity and cathepsin D is 280-fold more abundant in human brain than BACE 1. Nevertheless, as BACE 1 and cathepsin D show poor activity towards the WT beta-secretase site, they suggested continuing the search for additional beta-secretase(s). The second group reported cathepsin B as an alternative beta-secretase possessing excellent kinetic efficiency and specificity for the WT beta-secretase site. Significantly, inhibitors of cathepsin B improved memory, with reduced amyloid plaques and decreased Abeta(40/42) in brains of AD animal models expressing amyloid precursor protein containing the WT beta-secretase site. The third group addressed isoaspartate and pyroglutamate (pGlu) posttranslational modifications of Abeta. Results showed that cathepsin B, but not BACE 1, efficiently cleaves the WT beta-secretase isoaspartate site. Furthermore, cyclization of N-terminal Glu by glutaminyl cyclase generates highly amyloidogenic pGluAbeta(3-40/42). These presentations suggest cathepsin B and glutaminyl cyclase as potential new AD therapeutic targets.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/biosynthesis , Peptide Fragments/biosynthesis , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/genetics , Animals , Enzyme Inhibitors/therapeutic use , Humans , Peptide Fragments/genetics , Substrate Specificity
18.
Biol Chem ; 388(2): 247-52, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17261088

ABSTRACT

Abnormal accumulation of neurotoxic beta-amyloid peptides (Abeta) in brain represents a key factor in the progression of Alzheimer's disease (AD). Identification of small molecules that effectively reduce brain levels of Abeta is important for development of Abeta-lowering agents for AD. In this study, we demonstrate that in vivo Abeta levels in brain are significantly reduced by the cysteine protease inhibitor E64d and the related CA074Me inhibitor, which inhibits cathepsin B. Direct infusion of these inhibitors into brains of guinea pigs resulted in reduced levels of Abeta by 50-70% after 30 days of treatment. Substantial decreases in Abeta also occurred after only 7 days of inhibitor infusion, with a reduction in both Abeta40 and Abeta42 peptide forms. A prominent decrease in Abeta peptides was observed in brain synaptosomal nerve terminal preparations after CA074Me treatment. Analyses of APP-derived proteolytic fragments showed that CA074Me reduced brain levels of the CTFbeta fragment, and increased amounts of the sAPPalpha fragment. These results suggest that CA074Me inhibits Abeta production by modulating APP processing. Animals appeared healthy after treatment with these inhibitors. These results, showing highly effective in vivo decreases in brain Abeta levels by these cysteine protease inhibitors, indicate the feasibility of using related compounds for lowering Abeta in AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/drug effects , Brain/metabolism , Cysteine Endopeptidases/drug effects , Dipeptides/administration & dosage , Leucine/analogs & derivatives , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/drug effects , Animals , Cathepsin B/antagonists & inhibitors , Cysteine Endopeptidases/isolation & purification , Dipeptides/pharmacology , Guinea Pigs , Leucine/administration & dosage , Leucine/pharmacology , Molecular Weight , Protease Inhibitors/administration & dosage , Protease Inhibitors/pharmacology , Synaptosomes/chemistry
19.
Biol Chem ; 388(9): 979-83, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17696783

ABSTRACT

Beta-secretase inhibitors that lower brain beta-amyloid peptides (Abeta) are likely to be effective for treating Alzheimer's disease (AD). Irreversible epoxysuccinyl cysteine protease inhibitors are known to reduce brain Abeta and beta-secretase activity in the guinea pig model of human Abeta production. In this study, acetyl-L-leucyl-L-valyl-L-lysinal (Ac-LVK-CHO) is also shown to significantly reduce brain Abeta and beta-secretase activity and brain Abeta in the same model. Ac-LVK-CHO is structurally distinct from the epoxysuccinyl inhibitors and is a reversible cysteine protease inhibitor. The results suggest that cysteine protease inhibitors generally, and reversible cysteine protease inhibitors specifically, have potential for development as AD therapeutics.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Cathepsin B/metabolism , Cathepsins/metabolism , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Oligopeptides/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Animals , Brain/drug effects , Cathepsin L , Cysteine Proteinase Inhibitors/chemistry , Guinea Pigs , Humans , Oligopeptides/chemistry
20.
Biol Chem ; 386(9): 931-40, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16164418

ABSTRACT

The regulated secretory pathway of neurons is the major source of extracellular A beta that accumulates in Alzheimer's disease (AD). Extracellular A beta secreted from that pathway is generated by beta-secretase processing of amyloid precursor protein (APP). Previously, cysteine protease activity was demonstrated as the major beta-secretase activity in regulated secretory vesicles of neuronal chromaffin cells. In this study, the representative cysteine protease activity in these secretory vesicles was purified and identified as cathepsin B by peptide sequencing. Immunoelectron microscopy demonstrated colocalization of cathepsin B with A beta in these vesicles. The selective cathepsin B inhibitor, CA074, blocked the conversion of endogenous APP to A beta in isolated regulated secretory vesicles. In chromaffin cells, CA074Me (a cell permeable form of CA074) reduced by about 50% the extracellular A beta released by the regulated secretory pathway, but CA074Me had no effect on A beta released by the constitutive pathway. Furthermore, CA074Me inhibited processing of APP into the COOH-terminal beta-secretase-like cleavage product. These results provide evidence for cathepsin B as a candidate beta-secretase in regulated secretory vesicles of neuronal chromaffin cells. These findings implicate cathepsin B as beta-secretase in the regulated secretory pathway of brain neurons, suggesting that inhibitors of cathepsin B may be considered as therapeutic agents to reduce A beta in AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/biosynthesis , Cathepsin B/antagonists & inhibitors , Chromaffin Cells/metabolism , Endopeptidases/metabolism , Neurons/metabolism , Secretory Vesicles/metabolism , Alzheimer Disease/enzymology , Amino Acid Sequence , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Animals , Cathepsin B/isolation & purification , Cathepsin B/metabolism , Cattle , Chromaffin Cells/drug effects , Chromaffin Cells/enzymology , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Molecular Sequence Data , Neurons/drug effects , Neurons/enzymology , Secretory Vesicles/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL