Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Mol Basis Dis ; 1869(6): 166747, 2023 08.
Article in English | MEDLINE | ID: mdl-37207905

ABSTRACT

Neovascular inflammatory vitreoretinopathy (NIV) is a rare eye disease that ultimately leads to complete blindness and is caused by mutations in the gene encoding calpain-5 (CAPN5), with six pathogenic mutations identified. In transfected SH-SY5Y cells, five of the mutations resulted in decreased membrane association, diminished S-acylation, and reduced calcium-induced autoproteolysis of CAPN5. CAPN5 proteolysis of the autoimmune regulator AIRE was impacted by several NIV mutations. R243, L244, K250 and the adjacent V249 are on ß-strands in the protease core 2 domain. Conformational changes induced by Ca2+binding result in these ß-strands forming a ß-sheet and a hydrophobic pocket which docks W286 side chain away from the catalytic cleft, enabling calpain activation based on comparison with the Ca2+-bound CAPN1 protease core. The pathologic variants R243L, L244P, K250N, and R289W are predicted to disrupt the ß-strands, ß-sheet, and hydrophobic pocket, impairing calpain activation. The mechanism by which these variants impair membrane association is unclear. G376S impacts a conserved residue in the CBSW domain and is predicted to disrupt a loop containing acidic residues which may contribute to membrane binding. G267S did not impair membrane association and resulted in a slight but significant increase in autoproteolytic and proteolytic activity. However, G267S is also identified in individuals without NIV. Combined with the autosomal dominant pattern of NIV inheritance and evidence that CAPN5 may dimerize, the results are consistent with a dominant negative mechanism for the five pathogenic variants which resulted in impaired CAPN5 activity and membrane association and a gain-of-function for the G267S variant.


Subject(s)
Neuroblastoma , Vitreoretinopathy, Proliferative , Humans , Calpain/genetics , Calpain/metabolism , Vitreoretinopathy, Proliferative/genetics , Vitreoretinopathy, Proliferative/pathology , Mutation
2.
Biochim Biophys Acta Mol Cell Res ; 1869(9): 119298, 2022 09.
Article in English | MEDLINE | ID: mdl-35643222

ABSTRACT

Calpain-5 (CAPN5) is a member of the calpain family of calcium-activated neutral thiol proteases. CAPN5 is partly membrane associated, despite its lack of a transmembrane domain. Unlike classical calpains, CAPN5 contains a C-terminal C2 domain. C2 domains often have affinity to lipids, mediating membrane association. We recently reported that the C2 domain of CAPN5 was essential for its membrane association and the activation of its autolytic activity. However, despite the removal of the C2 domain by autolysis, the N-terminal fragment of CAPN5 remained membrane associated. S-acylation, also referred to as S-palmitoylation, is a reversible post-translational lipid modification of cysteine residues that promotes membrane association of soluble proteins. In the present study several S-acylated cysteine residues were identified in CAPN5 with the acyl-PEG exchange method. Data reported here demonstrate that CAPN5 is S-acylated on up to three cysteine residues including Cys-4 and Cys-512, and likely Cys-507. The D589N mutation in a potential calcium binding loop within the C2 domain interfered with the S-acylation of CAPN5, likely preventing initial membrane association. Mutating specific cysteine residues of CAPN5 interfered with both its membrane association and the activation of CAPN5 autolysis. Taken together, our results suggest that the S-acylation of CAPN5 is critical for its membrane localization which appears to favor its enzymatic activity.


Subject(s)
Calpain , Cysteine , Acylation , Calcium/metabolism , Calpain/genetics , Calpain/metabolism , Cysteine/genetics , Cysteine/metabolism , Lipoylation
3.
Biochim Biophys Acta Mol Cell Res ; 1868(7): 119019, 2021 06.
Article in English | MEDLINE | ID: mdl-33811937

ABSTRACT

The enzymatic characteristics of the ubiquitous calpain 5 (CAPN5) remain undescribed despite its high expression in the central nervous system and links to eye development and disease. CAPN5 contains the typical protease core domains but lacks the C terminal penta-EF hand domain of classical calpains, and instead contains a putative C2 domain. This study used the SH-SY5Y neuroblastoma cell line stably transfected with CAPN5-3xFLAG variants to assess the potential roles of the CAPN5 C2 domain in Ca2+ regulated enzyme activity and intracellular localization. Calcium dependent autoproteolysis of CAPN5 was documented and characterized. Mutation of the catalytic Cys81 to Ala or addition of EGTA prevented autolysis. Eighty µM Ca2+ was sufficient to stimulate half-maximal CAPN5 autolysis in cellular lysates. CAPN5 autolysis was inhibited by tri-leucine peptidyl aldehydes, but less effectively by di-Leu aldehydes, consistent with a more open conformation of the protease core relative to classical calpains. In silico modeling revealed a type II topology C2 domain including loops with the potential to bind calcium. Mutation of the acidic amino acid residues predicted to participate in Ca2+ binding, particularly Asp531 and Asp589, resulted in a decrease of CAPN5 membrane association. These residues were also found to be invariant in several genomes. The autolytic fragment of CAPN5 was prevalent in membrane-enriched fractions, but not in cytosolic fractions, suggesting that membrane association facilitates the autoproteolytic activity of CAPN5. Together, these results demonstrate that CAPN5 undergoes Ca2+-activated autoproteolytic processing and suggest that CAPN5 association with membranes enhances CAPN5 autolysis.


Subject(s)
C2 Domains/physiology , Calpain/genetics , Calpain/metabolism , Amino Acid Sequence/genetics , C2 Domains/genetics , Cell Movement , Enzyme Activation/genetics , Humans , Models, Molecular , Mutation/genetics , Protein Conformation , Protein Domains/physiology
4.
Int J Mol Sci ; 23(1)2021 Dec 29.
Article in English | MEDLINE | ID: mdl-35008785

ABSTRACT

Microglia/astrocyte and B cell neuroimmune responses are major contributors to the neurological deficits after traumatic spinal cord injury (SCI). Bruton tyrosine kinase (BTK) activation mechanistically links these neuroimmune mechanisms. Our objective is to use Ibrutinib, an FDA-approved BTK inhibitor, to inhibit the neuroimmune cascade thereby improving locomotor recovery after SCI. Rat models of contusive SCI, Western blot, immunofluorescence staining imaging, flow cytometry analysis, histological staining, and behavioral assessment were used to evaluate BTK activity, neuroimmune cascades, and functional outcomes. Both BTK expression and phosphorylation were increased at the lesion site at 2, 7, 14, and 28 days after SCI. Ibrutinib treatment (6 mg/kg/day, IP, starting 3 h post-injury for 7 or 14 days) reduced BTK activation and total BTK levels, attenuated the injury-induced elevations in Iba1, GFAP, CD138, and IgG at 7 or 14 days post-injury without reduction in CD45RA B cells, improved locomotor function (BBB scores), and resulted in a significant reduction in lesion volume and significant improvement in tissue-sparing 11 weeks post-injury. These results indicate that Ibrutinib exhibits neuroprotective effects by blocking excessive neuroimmune responses through BTK-mediated microglia/astroglial activation and B cell/antibody response in rat models of SCI. These data identify BTK as a potential therapeutic target for SCI.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Neuroimmunomodulation , Recovery of Function , Spinal Cord Injuries/immunology , Adenine/analogs & derivatives , Adenine/pharmacology , Adenine/therapeutic use , Agammaglobulinaemia Tyrosine Kinase/metabolism , Animals , Antibody Formation/drug effects , Astrocytes/pathology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Body Weight/drug effects , Calcium-Binding Proteins/metabolism , Glial Fibrillary Acidic Protein/metabolism , Immunoglobulin G/metabolism , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/pathology , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/pathology , Motor Activity/drug effects , Neuroimmunomodulation/drug effects , Phosphorylation/drug effects , Piperidines/pharmacology , Piperidines/therapeutic use , Plasma Cells/drug effects , Plasma Cells/metabolism , Rats , Recovery of Function/drug effects , Spinal Cord/drug effects , Spinal Cord/pathology , Spinal Cord/physiopathology , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Spleen/pathology , Syndecan-1/metabolism , Treatment Outcome , Up-Regulation/drug effects
5.
J Neurotrauma ; 37(21): 2268-2276, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32718209

ABSTRACT

Small molecule inhibitors of calcium-dependent proteases, calpains (CAPNs), protect against neurodegeneration induced by a variety of insults including excitotoxicity and spinal cord injury (SCI). Many of these compounds, however, also inhibit other proteases, which has made it difficult to evaluate the contribution of calpains to neurodegeneration. Calpastatin is a highly specific endogenous inhibitor of classical calpains, including CAPN1 and CAPN2. In the present study, we utilized transgenic mice that overexpress human calpastatin under the prion promoter (PrP-hCAST) to evaluate the hypothesis that calpastatin overexpression protects against excitotoxic hippocampal injury and contusive SCI. The PrP-hCAST organotypic hippocampal slice cultures showed reduced neuronal death and reduced calpain-dependent proteolysis (α-spectrin breakdown production, 145 kDa) at 24 h after N-methyl-D-aspartate (NMDA) injury compared with the wild-type (WT) cultures (n = 5, p < 0.05). The PrP-hCAST mice (n = 13) displayed a significant improvement in locomotor function at one and three weeks after contusive SCI compared with the WT controls (n = 9, p < 0.05). Histological assessment of lesion volume and tissue sparing, performed on the same animals used for behavioral analysis, revealed that calpastatin overexpression resulted in a 30% decrease in lesion volume (p < 0.05) and significant increases in tissue sparing, white matter sparing, and gray matter sparing at four weeks post-injury compared with WT animals. Calpastatin overexpression reduced α-spectrin breakdown by 51% at 24 h post-injury, compared with WT controls (p < 0.05, n = 3/group). These results provide support for the hypothesis that sustained calpain-dependent proteolysis contributes to pathological deficits after excitotoxic injury and traumatic SCI.


Subject(s)
Calcium-Binding Proteins/metabolism , Hippocampus/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Animals , Hippocampus/pathology , Humans , Locomotion/physiology , Mice , Mice, Transgenic
6.
J Neurotrauma ; 36(18): 2618-2630, 2019 09 15.
Article in English | MEDLINE | ID: mdl-30747048

ABSTRACT

We previously reported the serendipitous observation that fenbendazole, a benzimidazole anthelmintic, improved functional and pathological outcomes following thoracic spinal cord contusion injury in mice when administered pre-injury. Fenbendazole is widely used in veterinary medicine. However, it is not approved for human use and it was uncertain if only post-injury administration would offer similar benefits. In the present study we evaluated post-injury administration of a closely related, human anthelmintic drug, flubendazole, using a rat spinal cord contusion injury model. Flubendazole, administered i.p. 5 or 10 mg/kg day, beginning 3 h post-injury and daily thereafter for 2 or 4 weeks, resulted in improved locomotor function after contusion spinal cord injury (SCI) compared with vehicle-treated controls. Histological analysis of spinal cord sections showed that such treatment with flubendazole also reduced lesion volume and improved total tissue sparing, white matter sparing, and gray matter sparing. Flubendazole inhibited the activation of glial fibrillary acidic protein (GFAP); suppressed cyclin B1 expression and Bruton tyrosine kinase activation, markers of B cell activation/proliferation and inflammation; and reduced B cell autoimmune response. Together, these results suggest the use of the benzimidazole anthelmintic flubendazole as a potential therapeutic for SCI.


Subject(s)
Mebendazole/analogs & derivatives , Neuroprotective Agents/pharmacology , Recovery of Function/drug effects , Spinal Cord Injuries/pathology , Spinal Cord/drug effects , Animals , Antinematodal Agents/pharmacology , Drug Repositioning , Female , Mebendazole/pharmacology , Rats , Rats, Sprague-Dawley , Spinal Cord/pathology , Spinal Cord Injuries/physiopathology
7.
J Neurosci Methods ; 272: 33-37, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27168498

ABSTRACT

BACKGROUND: The extent and severity of traumatic brain injuries (TBIs) can be difficult to determine with current diagnostic methods. To address this, there has been increased interest in developing biomarkers to assist in the diagnosis, determination of injury severity, evaluation of recovery and therapeutic efficacy, and prediction of outcomes. Several promising serum TBI biomarkers have been identified using hypothesis-driven approaches, largely examining proteins that are abundant in neurons and non-neural cells in the CNS. NEW METHOD: An unbiased approach, phage display, was used to identify serum TBI biomarkers. In this proof-of-concept study, mice received a TBI using the controlled cortical impact model of TBI (1mm injury depth, 3.5m/s velocity) and phage display was utilized to identify putative serum biomarkers at 6h postinjury. RESULTS: An engineered phage which preferentially bound to injured serum was sequenced to identify the 12-mer 'recognizer' peptide expressed on the coat protein. Following synthesis of the recognizer peptide, pull down, and mass spectrometry analysis, the target protein was identified as glial fibrillary acidic protein (GFAP). COMPARISON WITH EXISTING METHODS AND CONCLUSIONS: GFAP has previously been identified as a promising TBI biomarker. The results provide proof of concept regarding the ability of phage display to identify TBI serum biomarkers. This methodology is currently being applied to serum biomarkers of mild TBI.


Subject(s)
Bacteriophages , Blood Chemical Analysis/methods , Brain Injuries, Traumatic/blood , Brain Injuries, Traumatic/diagnosis , Cell Surface Display Techniques , Glial Fibrillary Acidic Protein/blood , Amino Acid Sequence , Animals , Bacteriophages/genetics , Bacteriophages/metabolism , Biomarkers/blood , Capsid Proteins/genetics , Capsid Proteins/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Male , Mice, Inbred C57BL , Parietal Lobe , Peptide Library
8.
Invest Ophthalmol Vis Sci ; 57(6): 2509-21, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27152965

ABSTRACT

PURPOSE: We characterize calpain-5 (CAPN5) expression in retinal and neuronal subcellular compartments. METHODS: CAPN5 gene variants were classified using the exome variant server, and RNA-sequencing was used to compare expression of CAPN5 mRNA in the mouse and human retina and in retinoblastoma cells. Expression of CAPN5 protein was ascertained in humans and mice in silico, in mouse retina by immunohistochemistry, and in neuronal cancer cell lines and fractionated central nervous system tissue extracts by Western analysis with eight antibodies targeting different CAPN5 regions. RESULTS: Most CAPN5 genetic variation occurs outside its protease core; and searches of cancer and epilepsy/autism genetic databases found no variants similar to hyperactivating retinal disease alleles. The mouse retina expressed one transcript for CAPN5 plus those of nine other calpains, similar to the human retina. In Y79 retinoblastoma cells, the level of CAPN5 transcript was very low. Immunohistochemistry detected CAPN5 expression in the inner and outer nuclear layers and at synapses in the outer plexiform layer. Western analysis of fractionated retinal extracts confirmed CAPN5 synapse localization. Western blots of fractionated brain neuronal extracts revealed distinct subcellular patterns and the potential presence of autoproteolytic CAPN5 domains. CONCLUSIONS: CAPN5 is moderately expressed in the retina and, despite higher expression in other tissues, hyperactive disease mutants of CAPN5 only manifest as eye disease. At the cellular level, CAPN5 is expressed in several different functional compartments. CAPN5 localization at the photoreceptor synapse and with mitochondria explains the neural circuitry phenotype in human CAPN5 disease alleles.


Subject(s)
Calpain/genetics , Gene Expression Regulation, Neoplastic , Photoreceptor Cells/metabolism , RNA, Neoplasm/genetics , Retinal Neoplasms/genetics , Retinoblastoma/genetics , Synapses/metabolism , Animals , Blotting, Western , Calpain/biosynthesis , Cattle , Female , Humans , Immunohistochemistry , Male , Mice , Neoplasms, Experimental , Photoreceptor Cells/pathology , Retina/metabolism , Retina/pathology , Retinal Neoplasms/metabolism , Retinal Neoplasms/pathology , Retinoblastoma/metabolism , Retinoblastoma/pathology , Tumor Cells, Cultured
9.
J Biol Chem ; 289(28): 19383-94, 2014 Jul 11.
Article in English | MEDLINE | ID: mdl-24838245

ABSTRACT

Calpain 5 (CAPN5) is a non-classical member of the calpain family. It lacks the EF hand motif characteristic of classical calpains but retains catalytic and Ca(2+) binding domains, and it contains a unique C-terminal domain. TRA-3, an ortholog of CAPN5, has been shown to be involved in necrotic cell death in Caenorhabditis elegans. CAPN5 is expressed throughout the CNS, but its expression relative to other calpains and subcellular distribution has not been investigated previously. Based on relative mRNA levels, Capn5 is the second most highly expressed calpain in the rat CNS, with Capn2 mRNA being the most abundant. Unlike classical calpains, CAPN5 is a non-cytosolic protein localized to the nucleus and extra-nuclear locations. CAPN5 possesses two nuclear localization signals (NLS): an N-terminal monopartite NLS and a unique bipartite NLS closer to the C terminus. The C-terminal NLS contains a SUMO-interacting motif that contributes to nuclear localization, and mutation or deletion of both NLS renders CAPN5 exclusively cytosolic. Dual NLS motifs are common among transcription factors. Interestingly, CAPN5 is found in punctate domains associated with promyelocytic leukemia (PML) protein within the nucleus. PML nuclear bodies are implicated in transcriptional regulation, cell differentiation, cellular response to stress, viral defense, apoptosis, and cell senescence as well as protein sequestration, modification, and degradation. The roles of nuclear CAPN5 remain to be determined.


Subject(s)
Calpain/biosynthesis , Cell Nucleus/enzymology , Central Nervous System/metabolism , Gene Expression Regulation, Enzymologic/physiology , Intranuclear Inclusion Bodies/enzymology , Nuclear Localization Signals/metabolism , Amino Acid Motifs , Animals , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/biosynthesis , Caenorhabditis elegans Proteins/genetics , Calpain/genetics , Cell Nucleus/genetics , Intranuclear Inclusion Bodies/genetics , Male , Mice , Mice, Transgenic , Nuclear Localization Signals/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein , Rats , Rats, Sprague-Dawley , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
10.
Mol Neurobiol ; 49(3): 1282-92, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24297323

ABSTRACT

Excess glutamatergic neurotransmission may contribute to excitotoxic loss of nigrostriatal neurons in Parkinson's disease (PD). Here, we determined if increasing glutamate uptake could reduce the extent of tyrosine hydroxylase (TH) loss in PD progression. The beta-lactam antibiotic, ceftriaxone, increases the expression of glutamate transporter 1 (GLT-1), a glutamate transporter that plays a major role in glutamate clearance in central nervous system and may attenuate adverse behavioral or neurobiological function in other neurodegenerative disease models. In association with >80% TH loss, we observed a significant decrease in glutamate uptake in the established 6-hydroxydopamine (6-OHDA) PD model. Ceftriaxone (200 mg/kg, i.p.) increased striatal glutamate uptake with >5 consecutive days of injection in nonlesioned rats and lasted out to 14 days postinjection, a time beyond that required for 6-OHDA to produce >70% TH loss (∼9 days). When ceftriaxone was given at the time of 6-OHDA, TH loss was ∼57% compared to ∼85% in temporally matched vehicle-injected controls and amphetamine-induced rotation was reduced about 2-fold. This attenuation of TH loss was associated with increased glutamate uptake, increased GLT-1 expression, and reduced Serine 19 TH phosphorylation, a calcium-dependent target specific for nigrostriatal neurons. These results reveal that glutamate uptake can be targeted in a PD model, decrease the rate of TH loss in a calcium-dependent manner, and attenuate locomotor behavior associated with 6-OHDA lesion. Given that detection of reliable PD markers will eventually be employed in susceptible populations, our results give credence to the possibility that increasing glutamate uptake may prolong the time period before locomotor impairment occurs.


Subject(s)
Ceftriaxone/therapeutic use , Corpus Striatum/metabolism , Glutamic Acid/metabolism , Oxidopamine/toxicity , Parkinson Disease, Secondary/metabolism , Tyrosine 3-Monooxygenase/metabolism , Animals , Ceftriaxone/pharmacology , Corpus Striatum/drug effects , Male , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/drug therapy , Rats , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase/antagonists & inhibitors
11.
Exp Neurol ; 220(2): 316-9, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19751724

ABSTRACT

Calpains and caspases are cysteine endopeptidases which share many similar substrates. Caspases are essential for caspase-dependent apoptotic death where calpains may play an augmentive role, while calpains are strongly implicated in necrotic cell death morphologies. Previous studies have demonstrated a down-regulation in the expression of many components of the caspase-dependent cell death pathway during CNS development. We therefore sought to determine if there is a corresponding upregulation of calpains. The major CNS calpains are the mu-and m-isoforms, composed of the unique 80 kDa calpain 1 and 2 subunits, respectively, and the shared 28 kDa small subunit. In rat brain, relative protein and mRNA levels of calpain 1, calpain 2, caspase 3, and the endogenous calpain inhibitor-calpastatin, were evaluated using western blot and real-time RT-PCR. The developmental time points examined ranged from embryonic day 18 until postnatal day 90. Calpain 1 and calpastatin protein and mRNA levels were low at early developmental time points and increased dramatically by P30. Conversely, caspase-3 expression was greatest at E18, and was rapidly downregulated by P30. Calpain 2 protein and mRNA levels were relatively constant throughout the E18-P90 age range examined. The inverse relationship of calpain 1 and caspase 3 levels during CNS development is consistent with the shift from caspase-dependent to caspase-independent cell death mechanisms following CNS injury in neonatal vs. adult rat brain.


Subject(s)
Brain Chemistry/physiology , Brain/growth & development , Calcium-Binding Proteins/biosynthesis , Calpain/biosynthesis , Aging/metabolism , Animals , Animals, Newborn , Blotting, Western , Caspase 3/biosynthesis , Mitochondria/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
12.
Exp Neurol ; 218(2): 221-7, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19393648

ABSTRACT

Caspase-independent cell death, an important death pathway in many cells including neurons, is executed via apoptosis-inducing factor (AIF), an oxidoreductase, localized to the mitochondrial intermembrane space. AIF is processed and released from mitochondria following mitochondrial permeability transition pore (mPTP) formation, and translocates to the nucleus to induce DNA fragmentation and cell death. The release of AIF requires cleavage of its N-terminus anchored in the inner mitochondrial membrane. The protease responsible for this AIF truncation has not been established, although there is considerable evidence suggesting a role for micro-calpain. We previously found that a pool of micro-calpain is localized to the mitochondrial intermembrane space, the submitochondrial compartment in which AIF truncation occurs. The close submitochondrial proximity of mitochondrial micro-calpain and AIF gives support to the hypothesis that mitochondrial micro-calpain may be the protease responsible for processing AIF prior to its release. In the present study, AIF was released from rat liver mitochondria following mPTP induction by atractyloside. This release was inhibited by the cysteine protease inhibitor MDL28170, but not by more specific calpain inhibitors PD150606 and calpastatin. Atractyloside caused swelling in rat brain mitochondria, but did not induce AIF release. In a mitochondrial fraction from SH-SY5Y neuroblastoma cells, incubation with 5 mM Ca(2+) resulted in the activation of micro-calpain but not in AIF truncation. In summary, the localization of micro-calpain to the mitochondrial intermembrane space is suggestive of its possible involvement in AIF processing, but direct experimental evidence supporting such a role has been elusive.


Subject(s)
Apoptosis Inducing Factor/metabolism , Calpain/metabolism , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Swelling/drug effects , Acrylates/pharmacology , Animals , Atractyloside/pharmacology , Blotting, Western , Calcium/pharmacology , Calcium-Binding Proteins/pharmacology , Calpain/antagonists & inhibitors , Cell Line, Tumor , Cerebral Cortex/cytology , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Enzyme Inhibitors/pharmacology , Liver/cytology , Male , Mitochondria/drug effects , Mitochondrial Permeability Transition Pore , Rats , Rats, Sprague-Dawley
13.
J Biol Chem ; 283(6): 3409-3417, 2008 Feb 08.
Article in English | MEDLINE | ID: mdl-18070881

ABSTRACT

The ubiquitous m- and mu-calpains are thought to be localized in the cytosolic compartment, as is their endogenous inhibitor calpastatin. Previously, mu-calpain was found to be enriched in mitochondrial fractions isolated from rat cerebral cortex and SH-SY5Y neuroblastoma cells, but the submitochondrial localization of mu-calpain was not determined. In the present study, submitochondrial fractionation and digitonin permeabilization studies indicated that both calpain 1 and calpain small subunit 1, which together form mu-calpain, are present in the mitochondrial intermembrane space. The N terminus of calpain 1 contains an amphipathic alpha-helical domain, and is distinct from the N terminus of calpain 2. Calpain 1, but not calpain 2, was imported into mitochondria. Removal of the N-terminal 22 amino acids of calpain 1 blocked the mitochondrial calpain import, while addition of this N-terminal region to calpain 2 or green fluorescent protein enabled mitochondrial import. The N terminus of calpain 1 was not processed following mitochondrial import, but was removed by autolysis following calpain activation. Calpain small subunit 1 was not directly imported into mitochondria, but was imported in the presence of calpain 1. The presence of a mitochondrial targeting sequence in the N-terminal region of calpain 1 is consistent with the localization of mu-calpain to the mitochondrial intermembrane space and provides new insight into the possible functions of this cysteine protease.


Subject(s)
Calpain/chemistry , Gene Expression Regulation , Mitochondria/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Cerebral Cortex/metabolism , Cytosol/metabolism , Humans , Molecular Sequence Data , Protein Structure, Tertiary , Rats , Sequence Homology, Amino Acid , Subcellular Fractions/metabolism
14.
Biochem Biophys Res Commun ; 338(2): 1241-7, 2005 Dec 16.
Article in English | MEDLINE | ID: mdl-16259951

ABSTRACT

Calcium-dependent cysteine proteases, calpains, have physiological roles in cell motility and differentiation but also play a pathological role following insult or disease. The ubiquitous calpains are widely considered to be cytosolic enzymes, although there has been speculation of a mitochondrial calpain. Within a highly enriched fraction of mitochondria obtained from rat cortex and SH-SY5Y human neuroblastoma cells, immunoblotting demonstrated enrichment of the 80kDa mu-calpain large subunit and 28kDa small subunit. In rat cortex, antibodies against domains II and III of the large mu-calpain subunit also detected a 40kDa fragment, similar to the autolytic fragment generated following incubation of human erythrocyte mu-calpain with Ca(2+). Mitochondrial proteins including apoptosis inducing factor and mitochondrial Bax are calpain substrates, but the mechanism by which calpains gain access to these proteins is uncertain. Mitochondrial localization of mu-calpain places the enzyme in proximity to its mitochondrial substrates and to Ca(2+) released from mitochondrial stores.


Subject(s)
Calpain/metabolism , Mitochondria/metabolism , Neuroblastoma/metabolism , Animals , Cell Line, Tumor , Humans , Male , Rats , Rats, Sprague-Dawley , Tissue Distribution
15.
J Alzheimers Dis ; 7(1): 15-24, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15750211

ABSTRACT

Impaired tau catabolism may contribute to tau accumulation and aggregation in Alzheimer's disease and neurofibrillary tangle formation. This study examined the effects of proteasome and calpain inhibition on tau levels and turnover in primary rat hippocampal neurons and differentiated SH-SY5Y human neuroblastoma cells. Administration of proteasome (MG-115, lactacystin) or calpain (MDL28170) inhibitors for up to 24 hours did not alter tau levels in differentiated SH-SY5Y cells or rat hippocampal neurons. Addition of 1 microM and 10 microM MG-115 did not change total tau levels, but did result in increased reactivity of phosphorylation-dependent tau antibodies (PHF-1, CP-13) and decreased Tau-1 immunoreactivity. Administration of cycloheximide to inhibit de novo protein synthesis also did not alter tau levels in the presence or absence of lactacystin. These results demonstrate that although the proteasome and calpain protease systems are capable of degrading tau in cell-free assays, their inhibition does not alter cellular tau levels in primary neurons or differentiated neuroblastoma cells.


Subject(s)
Calpain/metabolism , Neural Inhibition/physiology , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neurons/metabolism , Neurons/pathology , Proteasome Endopeptidase Complex/metabolism , tau Proteins/metabolism , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Aminopeptidases/metabolism , Animals , Antibodies, Monoclonal/immunology , Cell Aggregation , Hippocampus/immunology , Hippocampus/metabolism , Hippocampus/pathology , In Vitro Techniques , Neuroblastoma/enzymology , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Neurons/immunology , Rats , Tumor Cells, Cultured , tau Proteins/immunology
16.
Exp Neurol ; 188(1): 161-70, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15191812

ABSTRACT

Excessive activation of calpains (calcium-activated neutral proteases) is observed following spinal cord contusion injury, traumatic brain injury, stroke, and in neurodegenerative disorders including Alzheimer's disease. Calpain inhibition represents an attractive therapeutic target, but current calpain inhibitors possess relatively weak potency, poor specificity, and in many cases, limited cellular and blood-brain barrier permeability. We developed novel calpain inhibitors consisting of the endogenous inhibitor, calpastatin or its inhibitory domain I, fused to the protein transduction domain of the HIV trans-activator (Tat) protein (Tat(47-57)). The Tat-calpastatin fusion proteins were potent calpain inhibitors in a cell-free activity assay, but did not inhibit cellular calpain activity in primary rat cortical neurons when applied exogenously at concentrations up to 5 microM. The fusion proteins were able to transduce neurons, but were localized within endosome-like structures. A similar endosomal uptake was observed for Tat-GFP. Together, the results suggest that endosomal uptake of the Tat-calpastatin prevents its interaction with calpain in other cellular compartments. Endosomal uptake of proteins fused to the Tat protein transduction domain severely limits the applications of this methodology.


Subject(s)
Calcium-Binding Proteins/pharmacology , Calpain/drug effects , Cerebral Cortex/drug effects , Neurons/drug effects , Recombinant Fusion Proteins/pharmacology , Animals , Brain Diseases/drug therapy , Brain Diseases/metabolism , Brain Diseases/physiopathology , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Calpain/metabolism , Cell Compartmentation/drug effects , Cell Compartmentation/genetics , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Dose-Response Relationship, Drug , Endosomes/drug effects , Endosomes/metabolism , Fetus , Gene Products, tat/genetics , Gene Products, tat/metabolism , Humans , Nerve Degeneration/drug therapy , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Neurons/metabolism , Protein Structure, Tertiary/genetics , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transduction, Genetic/methods
17.
J Neuropathol Exp Neurol ; 62(6): 633-43, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12834108

ABSTRACT

3-Nitropropionic acid (3NP), an irreversible inhibitor of succinate dehydrogenase, has been used to model features of neurodegenerative disorders including Huntington disease, as well as acute neuronal insults such as cerebral ischemia. 3NP induces rapid necrosis and delayed apoptosis in primary cultures of rat hippocampal neurons. Low levels of extracellular glutamate shift the cell death mechanism to necrosis, whereas antagonism of NMDA receptors results in predominately apoptotic death. In the present study, the involvement of cysteine proteases in the morphologic and biochemical alterations accompanying 3NP-induced neuron death was investigated. Immunoblots of spectrin breakdown products indicated Ca(2+)-dependent cysteine protease (calpain) activation within the 8 hours of 3NP administration, whereas caspase-3 activation was not evident until 16 to 48 hours after treatment. The NMDA receptor antagonist MK-801 (dizocilpine) decreased 3NP-induced calpain activity, but did not alter caspase-3 activity. Similar to MK-801, calpain inhibitors (Z-Val-Phe.H and Z-Leu-Phe-CONHEt) shifted the cell death morphology towards apoptosis and delayed, but did not prevent, the 3NP-induced cell death. Together, the results indicate that following 3NP administration, increased calpain activity precedes caspase-3 activation, contributes to the necrotic morphology, and facilitates and accelerates the cell death.


Subject(s)
Calpain/pharmacology , Cell Death , Neurons/drug effects , Neurotoxins/toxicity , Propionates/toxicity , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/physiology , Calpain/antagonists & inhibitors , Caspases/metabolism , Cell Survival/drug effects , Cells, Cultured , Cysteine Proteinase Inhibitors , Dizocilpine Maleate/pharmacology , Drug Synergism , Embryo, Mammalian , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Immunoblotting/methods , Immunohistochemistry/methods , Necrosis , Neurons/pathology , Nitro Compounds , Oligopeptides/pharmacology , Rats , Spectrin/metabolism , Time Factors
18.
Neurochem Int ; 43(2): 89-99, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12620277

ABSTRACT

3-Nitropropionic acid (3NP), an irreversible inhibitor of succinate dehydrogenase, induces both rapid necrotic and slow apoptotic death in rat hippocampal neurons. Low levels of extracellular glutamate (10 microM) shift the 3NP-induced cell death mechanism to necrosis, while NMDA receptor blockade results in predominantly apoptotic death. In this study, we examined the 3NP-induced alterations in free cytosolic and mitochondrial calcium levels, ATP levels, mitochondrial membrane potential, and calpain and caspase activity, under conditions resulting in the activation of apoptotic and necrotic pathways. In the presence of 10 microM glutamate, 3NP administration resulted in a massive elevation in [Ca(2+)](c) and [Ca(2+)](m), decreased ATP, rapid mitochondrial membrane depolarization, and a rapid activation of calpain but not caspase activity. In the presence of the NMDA receptor antagonist MK-801, 3NP did not induce a significant elevation of [Ca(2+)](c) within the 24h time period examined, nor increase [Ca(2+)](m) within 1h. ATP was maintained at control levels during the first hour of treatment, but declined 64% by 16h. Calpain and caspase activity were first evident at 24h following 3NP administration. 3NP treatment alone resulted in a more rapid decline in ATP, more rapid calpain activation (within 8h), and elevated [Ca(2+)](m) as compared to the results obtained with added MK-801. Together, the results demonstrate that 3NP-induced necrotic neuron death is associated with a massive calcium influx through NMDA receptors, resulting in mitochondrial depolarization and calpain activation; while 3NP-induced apoptotic neuron death is not associated with significant elevations in [Ca(2+)](c), nor with early changes in [Ca(2+)](m), mitochondrial membrane potential, ATP levels, or calpain activity.


Subject(s)
Adenosine Triphosphate/metabolism , Calcium/metabolism , Cell Death/drug effects , Intracellular Membranes/physiology , Membrane Potentials/physiology , Mitochondria/physiology , Neurons/physiology , Neurotoxins/toxicity , Propionates/toxicity , Animals , Brain/cytology , Brain/embryology , Cytosol/physiology , Dizocilpine Maleate/pharmacology , Female , Fetus/physiology , Glutamic Acid/pharmacology , In Vitro Techniques , Intracellular Membranes/drug effects , Membrane Potentials/drug effects , Mitochondria/drug effects , Neurons/cytology , Nitro Compounds , Pregnancy , Rats , Rats, Sprague-Dawley
19.
J Neurotrauma ; 20(1): 59-67, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12614588

ABSTRACT

Calpains (calcium-activated cysteine proteases) are strongly implicated in the secondary damage that follows contusion injury to the spinal cord. Calpains are activated within a few minutes following injury and their elevated activity persists for 24 h, thereby providing a reasonable window of opportunity for postinjury inhibition. Previous studies demonstrated decreased axonal damage and neurofilament proteolysis with postinjury intravenous administration of relatively low concentrations of the calpain inhibitors leupeptin, E-64-D, and calpeptin. We sought to determine if conditions under which calpain inhibitors were administered in previous studies resulted in effective calpain inhibition, and to identify conditions that result in significant calpain inhibition following spinal cord injury. Contusive spinal cord injury was produced in female Long-Evans rats using the NYU impactor at the 12.5-25-mm height setting. The results demonstrate that intravenous administration of 1 mg/kg E-64-D or 250 micro g/kg calpeptin does not inhibit total calpain activity in the rat spinal cord, measured using a BODIPY-FL labeled casein assay. Intravenous administration of MDL28170 (20 mg/kg) resulted in mild calpain inhibition and a modest decrease in the proteolysis of calpain substrates alpha-spectrin and MAP2. Intraspinal microinjection of 50 nmoles/19 micro g MDL28170, either 30 min prior to or 20 min following contusion injury, resulted in a more robust inhibition of total calpain activity and greater attenuation of alpha-spectrin breakdown and MAP2 proteolysis. The decreased proteolysis persisted 24 h postinjury. Together, the results demonstrate that direct microinjection of the calpain inhibitor MDL28170 is more effective than intravenous infusion in reducing calpain activity and decreasing the injury-induced proteolysis of calpain substrates.


Subject(s)
Calpain/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/enzymology , Animals , Blotting, Western , Calpain/metabolism , Female , Microinjections , Microtubule-Associated Proteins/metabolism , Rats , Rats, Long-Evans , Spinal Cord/drug effects , Spinal Cord/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...