Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Traffic ; 13(9): 1261-72, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22672335

ABSTRACT

The receptor tyrosine kinase Met and its ligand, the hepatocyte growth factor/scatter factor, are essential for embryonic development, whereas deregulation of Met signaling pathways is associated with tumorigenesis and metastasis. The presenilin-regulated intramembrane proteolysis (PS-RIP) is involved in ligand-independent downregulation of Met. This proteolytic process involves shedding of the Met extracellular domain followed by γ-secretase cleavage, generating labile intracellular fragments degraded by the proteasome. We demonstrate here that upon shedding both generated Met N- and C-terminal fragments are degraded directly in the lysosome, with C-terminal fragments escaping γ-secretase cleavage. PS-RIP and lysosomal degradation are complementary, because their simultaneous inhibition induces synergistic accumulation of fragments. Met N-terminal fragments associate with the high-affinity domain of HGF/SF, confirming its decoy activity which could be reduced through their routing to the lysosome at the expense of extracellular release. Finally, the DN30 monoclonal antibody inducing Met shedding promotes receptor degradation through induction of both PS-RIP and the lysosomal pathway. Thus, we demonstrate that Met shedding initiates a novel lysosomal degradation which participates to ligand-independent downregulation of the receptor.


Subject(s)
Lysosomes/enzymology , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-met/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Animals , Cell Line, Tumor , Dogs , Hepatocyte Growth Factor/metabolism , Humans , Intracellular Membranes/metabolism , Mice , NIH 3T3 Cells , Presenilins/metabolism , Protease Inhibitors/pharmacology , Proteolysis , Proto-Oncogene Proteins c-met/genetics , RNA, Small Interfering
2.
J Biol Chem ; 288(28): 20378-91, 2013 Jul 12.
Article in English | MEDLINE | ID: mdl-23737521

ABSTRACT

Understanding the regulatory mechanisms mediating PRNP gene expression is highly relevant to elucidating normal cellular prion protein (PrP) function(s) and the transmissibility of prion protein neurodegenerative diseases. Here, luciferase reporter assays showed that an endoplasmic reticulum stress element (ERSE)-like element, CCAAT-N26-CCACG in the human PRNP promoter, is regulated by ER stress and X-box-binding protein 1 (XBP1) but not by activating transcription factor 6 α (ATF6α). Bioinformatics identified the ERSE-26 motif in 37 other human genes in the absence of canonical ERSE sites except for three genes. Several of these genes are associated with a synaptic function or are involved in oxidative stress. Brefeldin A, tunicamycin, and thapsigargin ER stressors induced gene expression of PRNP and four randomly chosen ERSE-26-containing genes, ERLEC1, GADD45B, SESN2, and SLC38A5, in primary human neuron cultures or in the breast carcinoma MCF-7 cell line, although the level of the response depends on the gene analyzed, the genetic background of the cells, the cell type, and the ER stressor. Overexpression of XBP1 increased, whereas siRNA knockdown of XBP1 considerably reduced, PRNP and ERLEC1 mRNA levels in MCF-7 cells. Taken together, these results identify a novel ER stress regulator, which implicates the ER stress response in previously unrecognized cellular functions.


Subject(s)
DNA-Binding Proteins/metabolism , Endoplasmic Reticulum Stress , Gene Expression Regulation , Response Elements , Transcription Factors/metabolism , Activating Transcription Factor 6/genetics , Activating Transcription Factor 6/metabolism , Amino Acid Transport Systems, Neutral/genetics , Antigens, Differentiation/genetics , Base Sequence , Blotting, Western , Brefeldin A/pharmacology , Cells, Cultured , DNA-Binding Proteins/genetics , HEK293 Cells , Humans , Lectins/genetics , MCF-7 Cells , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Nuclear Proteins/genetics , Nucleotide Motifs/genetics , Prion Proteins , Prions/genetics , Promoter Regions, Genetic/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Regulatory Factor X Transcription Factors , Reverse Transcriptase Polymerase Chain Reaction , Thapsigargin/pharmacology , Transcription Factors/genetics , Tunicamycin/pharmacology , X-Box Binding Protein 1
3.
J Biol Chem ; 287(8): 5366-78, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22184117

ABSTRACT

The tumor suppressor gene hypermethylated in cancer 1 (HIC1), which encodes a transcriptional repressor, is epigenetically silenced in many human tumors. Here, we show that ectopic expression of HIC1 in the highly malignant MDA-MB-231 breast cancer cell line severely impairs cell proliferation, migration, and invasion in vitro. In parallel, infection of breast cancer cell lines with a retrovirus expressing HIC1 also induces decreased mRNA and protein expression of the tyrosine kinase receptor EphA2. Moreover, chromatin immunoprecipitation (ChIP) and sequential ChIP experiments demonstrate that endogenous HIC1 proteins are bound, together with the MTA1 corepressor, to the EphA2 promoter in WI38 cells. Taken together, our results identify EphA2 as a new direct target gene of HIC1. Finally, we observe that inactivation of endogenous HIC1 through RNA interference in normal breast epithelial cells results in the up-regulation of EphA2 and is correlated with increased cellular migration. To conclude, our results involve the tumor suppressor HIC1 in the transcriptional regulation of the tyrosine kinase receptor EphA2, whose ligand ephrin-A1 is also a HIC1 target gene. Thus, loss of the regulation of this Eph pathway through HIC1 epigenetic silencing could be an important mechanism in the pathogenesis of epithelial cancers.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , Receptor, EphA2/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Down-Regulation/genetics , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Gene Silencing , HEK293 Cells , Histone Deacetylases/metabolism , Humans , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Promoter Regions, Genetic/genetics , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/metabolism , Trans-Activators
4.
J Biol Chem ; 287(8): 5379-89, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22194601

ABSTRACT

The transcriptional repressor HIC1 (Hypermethylated in Cancer 1) is a tumor suppressor gene inactivated in many human cancers including breast carcinomas. In this study, we show that HIC1 is a direct transcriptional repressor of ß-2 adrenergic receptor (ADRB2). Through promoter luciferase activity, chromatin immunoprecipitation (ChIP) and sequential ChIP experiments, we demonstrate that ADRB2 is a direct target gene of HIC1, endogenously in WI-38 cells and following HIC1 re-expression in breast cancer cells. Agonist-mediated stimulation of ADRB2 increases the migration and invasion of highly malignant MDA-MB-231 breast cancer cells but these effects are abolished following HIC1 re-expression or specific down-regulation of ADRB2 by siRNA treatment. Our results suggest that early inactivation of HIC1 in breast carcinomas could predispose to stress-induced metastasis through up-regulation of the ß-2 adrenergic receptor.


Subject(s)
Breast Neoplasms/pathology , Cell Movement , Kruppel-Like Transcription Factors/metabolism , Receptors, Adrenergic, beta-2/genetics , Stress, Physiological , Breast Neoplasms/genetics , Breast Neoplasms/physiopathology , Cell Adhesion/genetics , Cell Line, Tumor , Cell Movement/genetics , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Neoplasm Invasiveness , Neoplasm Metastasis , RNA Interference , RNA, Small Interfering/genetics , Receptors, Adrenergic, beta-2/deficiency , Stress, Physiological/genetics
5.
Cell Death Dis ; 12(3): 227, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33649324

ABSTRACT

Active Caspase-6 (Casp6) and Tau cleaved by Casp6 at amino acids 402 (Tau∆D402) and 421 (Tau∆D421) are present in early Alzheimer disease intraneuronal neurofibrillary tangles, which are made primarily of filamentous Tau aggregates. To assess whether Casp6 cleavage of Tau contributes to Tau pathology and Casp6-mediated age-dependent cognitive impairment, we generated transgenic knock-in mouse models that conditionally express full-length human Tau (hTau) 0N4R only (CTO) or together with human Casp6 (hCasp6) (CTC). Region-specific hippocampal and cortical hCasp6 and hTau expression were confirmed with western blot and immunohistochemistry in 2-25-month-old brains. Casp6 activity was confirmed with Tau∆D421 and Tubulin cleaved by Casp6 immunopositivity in 3-25-month-old CTC, but not in CTO, brains. Immunoprecipitated Tau∆D402 was detected in both CTC and CTO brains, but was more abundant in CTC brains. Intraneuronal hippocampal Tau hyperphosphorylation at S202/T205, S422, and T231, and Tau conformational change were absent in both CTC and CTO brains. A slight accumulation of Tau phosphorylated at S396/404 and S202 was observed in Cornu Ammonis 1 (CA1) hippocampal neuron soma of CTC compared to CTO brains. Eighteen-month-old CTC brains showed rare argentophilic deposits that increased by 25 months, whereas CTO brains only displayed them sparsely at 25 months. Tau microtubule binding was equivalent in CTC and CTO hippocampi. Episodic and spatial memory measured with novel object recognition and Barnes maze, respectively, remained normal in 3-25-month-old CTC and CTO mice, in contrast to previously observed impairments in ACL mice expressing equivalent levels of hCasp6 only. Consistently, the CTC and CTO hippocampal CA1 region displayed equivalent dendritic spine density and no glial inflammation. Together, these results reveal that active hCasp6 co-expression with hTau generates Tau cleavage and rare age-dependent argentophilic deposits but fails to induce cognitive deficits, neuroinflammation, and Tau pathology.


Subject(s)
Alzheimer Disease/enzymology , Behavior, Animal , Brain/enzymology , Caspase 6/metabolism , Cognition , Cognitive Dysfunction/enzymology , Nerve Degeneration , Neuroglia/enzymology , Neurons/enzymology , tau Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Animals , Brain/pathology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Caspase 6/genetics , Cognitive Dysfunction/genetics , Cognitive Dysfunction/pathology , Cognitive Dysfunction/psychology , Disease Models, Animal , Locomotion , Memory , Mice, Inbred C57BL , Mice, Transgenic , Neurofibrillary Tangles/enzymology , Neurofibrillary Tangles/genetics , Neurofibrillary Tangles/pathology , Neuroglia/pathology , Neurons/pathology , Open Field Test , Phosphorylation , Protein Aggregates , Protein Aggregation, Pathological , tau Proteins/genetics
6.
Nat Commun ; 11(1): 4571, 2020 09 11.
Article in English | MEDLINE | ID: mdl-32917871

ABSTRACT

Early therapeutic interventions are essential to prevent Alzheimer Disease (AD). The association of several inflammation-related genetic markers with AD and the early activation of pro-inflammatory pathways in AD suggest inflammation as a plausible therapeutic target. Inflammatory Caspase-1 has a significant impact on AD-like pathophysiology and Caspase-1 inhibitor, VX-765, reverses cognitive deficits in AD mouse models. Here, a one-month pre-symptomatic treatment of Swedish/Indiana mutant amyloid precursor protein (APPSw/Ind) J20 and wild-type mice with VX-765 delays both APPSw/Ind- and age-induced episodic and spatial memory deficits. VX-765 delays inflammation without considerably affecting soluble and aggregated amyloid beta peptide (Aß) levels. Episodic memory scores correlate negatively with microglial activation. These results suggest that Caspase-1-mediated inflammation occurs early in the disease and raise hope that VX-765, a previously Food and Drug Administration-approved drug for human CNS clinical trials, may be a useful drug to prevent the onset of cognitive deficits and brain inflammation in AD.


Subject(s)
Aging/metabolism , Alzheimer Disease/metabolism , Cognitive Dysfunction/metabolism , Serpins/metabolism , Viral Proteins/metabolism , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Animals , Behavior, Animal , Cognitive Dysfunction/drug therapy , Cytokines/metabolism , Dipeptides/blood , Dipeptides/pharmacology , Disease Models, Animal , Encephalitis/metabolism , Encephalitis/pathology , Female , Humans , Inflammation/metabolism , Male , Memory Disorders/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Serpins/blood , Serpins/pharmacology , Spatial Memory/physiology , Viral Proteins/blood , Viral Proteins/pharmacology , para-Aminobenzoates/blood , para-Aminobenzoates/pharmacology
7.
J Alzheimers Dis ; 67(3): 893-910, 2019.
Article in English | MEDLINE | ID: mdl-30689579

ABSTRACT

Alzheimer's disease (AD) occurs as either an autosomal dominant inherited disease or sporadically. While familial mutant genes can be expressed in cells or in animal models to assess dysregulated functions, sporadic AD cannot be replicated in models given our lack of understanding of causality. Furthermore, the study of sporadic forms of AD is difficult given the inaccessibility of brain tissues in living individuals and the manifestation of symptoms years after the onset of disease. Here, the objective was to assess if induced pluripotent stem cell-derived neurons from well-ascertained sporadic AD individuals could represent potential cellular models to determine the underlying molecular mechanisms of disease. We used cryopreserved peripheral blood mononuclear cells from three well-ascertained sporadic AD and three non-cognitively impaired (NCI) individuals of the CIMA-Q cohort to obtain iPSC-derived neurons. Microtubule associated protein 2 was decreased in AD neurons, whereas expression of AD-associated amyloid precursor protein, tau, and amyloid-ß peptide was similar in AD and NCI individuals. RNA sequencing identified several upregulated and downregulated mRNAs in AD relative to NCI neurons. Of these, complement Factor H (CFH), signal regulatory protein beta1 (SIRPB1), and insulin like growth factor binding protein 5 (IGFBP5) were previously associated with AD. In addition, several transcription factors not previously associated with AD, but involved in neuronal proliferation and differentiation were differentially expressed. The results identify novel avenues for the study of the underlying causes of sporadic AD and support the establishment of additional lines to identify mechanisms of disease in sporadic AD individuals.


Subject(s)
Alzheimer Disease/pathology , Induced Pluripotent Stem Cells/pathology , Neurons/pathology , Aged , Aged, 80 and over , Alzheimer Disease/etiology , Amyloid beta-Peptides/metabolism , Cell Line , Cholinergic Neurons/cytology , Cholinergic Neurons/metabolism , Cholinergic Neurons/pathology , Female , Fluorescent Antibody Technique , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Male , Neurons/cytology , Neurons/metabolism , RNA/genetics , Real-Time Polymerase Chain Reaction , Exome Sequencing , tau Proteins/metabolism
8.
Biochem Biophys Res Commun ; 367(3): 573-7, 2008 Mar 14.
Article in English | MEDLINE | ID: mdl-18187039

ABSTRACT

The MET tyrosine kinase receptor activated by its ligand HGF/SF, induces several cellular responses, including survival. Nonetheless, the MET receptor is cleaved in stress conditions by caspases within its intracellular region, generating a 40kDa fragment, p40 MET, with pro-apoptotic properties. Here, we established that this cleavage splits the receptor at the juxtamembrane ESVD site, causing the concomitant generation of p100 MET, corresponding to the entire extracellular region of the MET receptor still spanning the membrane. This fragment is able to bind HGF/SF and to prevent HGF-dependent signaling downstream of full MET, demonstrating its function as a decoy receptor.


Subject(s)
Caspases/metabolism , Hepatocyte Growth Factor/metabolism , Peptide Fragments/biosynthesis , Proto-Oncogene Proteins c-met/metabolism , Animals , Apoptosis/drug effects , Cell Fractionation , Cells, Cultured , Dogs , Gene Transfer Techniques , Hepatocyte Growth Factor/antagonists & inhibitors , Hepatocyte Growth Factor/chemistry , Humans , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Proto-Oncogene Proteins c-met/chemistry , Proto-Oncogene Proteins c-met/genetics , Signal Transduction/drug effects , TNF-Related Apoptosis-Inducing Ligand/pharmacology
9.
Nat Commun ; 9(1): 3916, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30254377

ABSTRACT

Alzheimer's disease (AD) is an intractable progressive neurodegenerative disease characterized by cognitive decline and dementia. An inflammatory neurodegenerative pathway, involving Caspase-1 activation, is associated with human age-dependent cognitive impairment and several classical AD brain pathologies. Here, we show that the nontoxic and blood-brain barrier permeable small molecule Caspase-1 inhibitor VX-765 dose-dependently reverses episodic and spatial memory impairment, and hyperactivity in the J20 mouse model of AD. Cessation of VX-765 results in the reappearance of memory deficits in the mice after 1 month and recommencement of treatment re-establishes normal cognition. VX-765 prevents progressive amyloid beta peptide deposition, reverses brain inflammation, and normalizes synaptophysin protein levels in mouse hippocampus. Consistent with these findings, Caspase-1 null J20 mice are protected from episodic and spatial memory deficits, neuroinflammation and Aß accumulation. These results provide in vivo proof of concept for Caspase-1 inhibition against AD cognitive deficits and pathologies.


Subject(s)
Alzheimer Disease/prevention & control , Caspase 1/metabolism , Caspase Inhibitors/pharmacology , Cognition Disorders/prevention & control , Disease Models, Animal , Memory Disorders/prevention & control , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/physiopathology , Caspase 1/genetics , Cognition/drug effects , Cognition/physiology , Cognition Disorders/metabolism , Cognition Disorders/physiopathology , Dipeptides/pharmacology , Humans , Memory/drug effects , Memory/physiology , Memory Disorders/metabolism , Memory Disorders/physiopathology , Mice, Knockout , para-Aminobenzoates/pharmacology
10.
Cell Death Differ ; 25(7): 1319-1335, 2018 07.
Article in English | MEDLINE | ID: mdl-29352267

ABSTRACT

Active cysteinyl protease Caspase-6 is associated with early Alzheimer and Huntington diseases. Higher entorhinal cortex and hippocampal Caspase-6 levels correlate with lower cognitive performance in aged humans. Caspase-6 induces axonal degeneration in human primary neuron cultures and causes inflammation and neurodegeneration in mouse hippocampus, and age-dependent memory impairment. To assess whether Caspase-6 causes damage to another neuronal system, a transgenic knock-in mouse overexpressing a self-activated form of Caspase-6 five-fold in the striatum, the area affected in Huntington disease, and 2.5-fold in the hippocampus and cortex, was generated. Detection of Tubulin cleaved by Caspase-6 confirmed Caspase-6 activity. The Caspase-6 expressing mice and control littermates were subjected to behavioral tests to assess Huntington disease-relevant psychiatric, motor, and cognitive deficits. Depression was excluded with the forced swim and sucrose consumption tests. Motor deficits were absent in the nesting, clasping, rotarod, vertical pole, gait, and open field analyzes. However, Caspase-6 mice developed age-dependent episodic and spatial memory deficits identified by novel object recognition, Barnes maze and Morris water maze assays. Neuron numbers were maintained in the striatum, hippocampus, and cortex. Microglia and astrocytes were increased in the hippocampal stratum lacunosum molecular and in the cortex, but not in the striatum. Synaptic mRNA profiling identified two differentially expressed genes in transgenic hippocampus, but none in striatum. Caspase-6 impaired synaptic transmission and induced neurodegeneration in hippocampal CA1 neurons, but not in striatal medium spiny neurons. These data revealed that active Caspase-6 in the striatal medium spiny neurons failed to induce inflammation, neurodegeneration or behavioral abnormalities, whereas active Caspase-6 in the cortex and hippocampus impaired episodic and spatial memories, and induced inflammation, neuronal dysfunction, and neurodegeneration. The results indicate age and neuronal subtype-dependent Caspase-6 toxicity and highlight the importance of targeting the correct neuronal subtype to identify underlying molecular mechanisms of neurodegenerative diseases.


Subject(s)
Caspase 6/metabolism , Cerebral Cortex/enzymology , Corpus Striatum/enzymology , Hippocampus/enzymology , Huntington Disease/enzymology , Memory Disorders/enzymology , Neurons/enzymology , Animals , Caspase 6/genetics , Cerebral Cortex/pathology , Corpus Striatum/pathology , Hippocampus/pathology , Humans , Huntington Disease/pathology , Memory Disorders/genetics , Memory Disorders/pathology , Mice , Mice, Transgenic , Neurons/pathology
11.
Ann N Y Acad Sci ; 1090: 188-202, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17384262

ABSTRACT

Hepatocyte growth factor/scatter factor (HGF/SF) induces scattering, morphogenesis, and survival of epithelial cells through activation of the MET tyrosine kinase receptor. HGF/SF and MET are involved in normal development and tumor progression of many tissues and organs, including the mammary gland. In order to find target genes of HGF/SF involved in its survival function, we used an oligonucleotide microarray representing 1,920 genes known to be involved in apoptosis, transcriptional regulation, and signal transduction. MCF-10A human mammary epithelial cells were grown in the absence of serum and treated or not with HGF/SF for 2 h. Total RNA was reverse-transcribed to cDNA in the presence of fluorescent Cy3-dUTP or Cy5-dUTP to generate fluorescently labeled cDNA probes. Microarrays were performed and the ratios of Cy5/Cy3 fluorescence were determined. The expression of three apoptotic genes was modified by HGF/SF, with A20 being upregulated, and DAXX and SMAC being downregulated. These changes of expression were confirmed by real-time quantitative PCR. According to current-knowledge, A20 is antiapoptotic and SMAC is proapoptotic, while a pro- or antiapoptotic function of DAXX is controversial. The fact that HGF/SF upregulates an antiapoptotic gene (A20) and downregulates a proapoptotic gene (SMAC) is in agreement with its survival effect in MCF-10A cells. This study identified novel apoptotic genes regulated by HGF/SF, which can contribute to its survival effect.


Subject(s)
Apoptosis/genetics , Gene Expression Regulation/physiology , Hepatocyte Growth Factor/physiology , Mammary Glands, Human/metabolism , Base Sequence , Blotting, Western , Cell Line , DNA Primers , Epithelial Cells/metabolism , Humans , Mammary Glands, Human/cytology , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
13.
Acta Neuropathol Commun ; 4(1): 127, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27931265

ABSTRACT

Abnormally elevated hippocampal Caspase-6 (Casp6) activity is intimately associated with age-related cognitive impairment in humans and in mice. In humans, these high levels of Casp6 activity are initially localized in the entorhinal cortex, the area of the brain first affected by the formation of neurofibrillary tangles, according to Braak staging. The reason for the high vulnerability of entorhinal cortex neurons to neurofibrillary tangle pathology and Casp6 activity is unknown. Casp6 activity is involved in axonal degeneration, therefore, one possibility to explain increased vulnerability of the entorhinal cortex neurons would be that the afferent neurons of the olfactory bulb, some of which project their axons to the entorhinal cortex, are equally degenerating. To examine this possibility, we examined the presence of Casp6 activity, neurofibrillary tangle formation and amyloid deposition by immunohistochemistry with neoepitope antisera against the p20 subunit of active Casp6 and Tau cleaved by Casp6 (Tau∆Casp6), phosphorylated Tau paired helical filament (PHF-1) antibodies and anti-ß-amyloid antiserum, respectively, in brains from individuals with no or mild cognitive impairment and Alzheimer disease (AD) dementia. Co-localization of Casp6 activity, PHF-1 and ß-amyloid was detected mostly in the anterior olfactory nucleus (AON) of the olfactory bulb. The levels of active Casp6 in the AON, which were the highest in the AD brains, correlated with PHF-1 levels, but not with ß-amyloid levels. AON Tau∆Casp6 levels correlated with entorhinal cortex Casp6 activity and PHF-1 levels. Multiple regression analyses demonstrated that AON Casp6 activity was associated with lower global cognitive function, mini mental state exam, episodic memory and semantic memory scores. These results suggest that AON Casp6 activity could lead to Casp6-mediated degeneration in the entorhinal cortex, but cannot exclude the possibilities that entorhinal cortex degeneration signals degeneration in the AON or that the pathologies occur in both regions independently. Nevertheless, AON Casp6 activity reflects that of the entorhinal cortex.


Subject(s)
Alzheimer Disease/enzymology , Caspase 6/metabolism , Cognitive Dysfunction/enzymology , Olfactory Bulb/enzymology , Olfactory Cortex/enzymology , Aged, 80 and over , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Cognitive Dysfunction/pathology , DNA-Binding Proteins/metabolism , Female , Hippocampus/metabolism , Hippocampus/pathology , Humans , Immunohistochemistry , Male , Memory/physiology , Mental Status Schedule , Neuropsychological Tests , Olfactory Bulb/pathology , Olfactory Cortex/pathology , Polycomb-Group Proteins/metabolism , Regression Analysis , Severity of Illness Index , tau Proteins/metabolism
14.
Sci Rep ; 5: 13730, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26400108

ABSTRACT

Methylene blue, currently in phase 3 clinical trials against Alzheimer Disease, disaggregates the Tau protein of neurofibrillary tangles by oxidizing specific cysteine residues. Here, we investigated if methylene blue can inhibit caspases via the oxidation of their active site cysteine. Methylene blue, and derivatives, azure A and azure B competitively inhibited recombinant Caspase-6 (Casp6), and inhibited Casp6 activity in transfected human colon carcinoma cells and in serum-deprived primary human neuron cultures. Methylene blue also inhibited recombinant Casp1 and Casp3. Furthermore, methylene blue inhibited Casp3 activity in an acute mouse model of liver toxicity. Mass spectrometry confirmed methylene blue and azure B oxidation of the catalytic Cys163 cysteine of Casp6. Together, these results show a novel inhibitory mechanism of caspases via sulfenation of the active site cysteine. These results indicate that methylene blue or its derivatives could (1) have an additional effect against Alzheimer Disease by inhibiting brain caspase activity, (2) be used as a drug to prevent caspase activation in other conditions, and (3) predispose chronically treated individuals to cancer via the inhibition of caspases.


Subject(s)
Caspase Inhibitors/pharmacology , Caspases/metabolism , Cysteine/metabolism , Methylene Blue/pharmacology , Oxidation-Reduction , Catalysis , Cell Line , Enzyme Activation/drug effects , Humans , Neurons/drug effects , Neurons/metabolism , Phenothiazines/pharmacology
15.
PLoS One ; 9(12): e114270, 2014.
Article in English | MEDLINE | ID: mdl-25470254

ABSTRACT

Caspases play an important role in maintaining tissue homeostasis. Active Caspase-6 (Casp6) is considered a novel therapeutic target against Alzheimer disease (AD) since it is present in AD pathological brain lesions, associated with age-dependent cognitive decline, and causes age-dependent cognitive impairment in the mouse brain. However, active Casp6 is highly expressed and activated in normal human colon epithelial cells raising concerns that inhibiting Casp6 in AD may promote colon carcinogenesis. Furthermore, others have reported rare mutations of Casp6 in human colorectal cancers and an effect of Casp6 on apoptosis and metastasis of colon cancer cell lines. Here, we investigated the role of Casp6 in inflammation-associated azoxymethane/dextran sulfate sodium (AOM/DSS) colon cancer in Casp6-overexpressing and -deficient mice. In wild-type mice, AOM/DSS-induced tumors had significantly higher Casp6 mRNA, protein and activity levels compared to normal adjacent colon tissues. Increased human Casp6 or absence of Casp6 expression in mice colon epithelial cells did not change colonic tumor multiplicity, burden or distribution. Nevertheless, the incidence of hyperplasia was slightly reduced in human Casp6-overexpressing colons and increased in Casp6 null colons. Overexpression of Casp6 did not affect the grade of the tumors while all tumors in heterozygous or homozygous Casp6 null colons were high grade compared to only 50% high grade in wild-type mice. Casp6 levels did not alter cellular proliferation and apoptosis. These results suggest that Casp6 is unlikely to be involved in colitis-associated tumors.


Subject(s)
Carcinogenesis/metabolism , Caspase 6/physiology , Colitis/enzymology , Colonic Neoplasms/enzymology , Animals , Apoptosis , Carcinogenesis/immunology , Cell Proliferation , Colitis/immunology , Colitis/pathology , Colon/enzymology , Colon/pathology , Colonic Neoplasms/etiology , Colonic Neoplasms/immunology , Female , Male , Mice, Inbred C57BL , Mice, Knockout
16.
PLoS One ; 8(11): e79313, 2013.
Article in English | MEDLINE | ID: mdl-24265764

ABSTRACT

Caspase-6 is an effector caspase that has not been investigated thoroughly despite the fact that Caspase-6 is strongly activated in Alzheimer disease brains. To understand the full physiological impact of Caspase-6 in humans, we investigated Caspase-6 expression. We performed western blot analyses to detect the pro-Caspase-6 and its active p20 subunit in fetal and adult lung, kidney, brain, spleen, muscle, stomach, colon, heart, liver, skin, and adrenals tissues. The levels were semi-quantitated by densitometry. The results show a ubiquitous expression of Caspase-6 in most fetal tissues with the lowest levels in the brain and the highest levels in the gastrointestinal system. Caspase-6 active p20 subunits were only detected in fetal stomach. Immunohistochemical analysis of a human fetal embryo showed active Caspase-6 positive apoptotic cells in the dorsal root ganglion, liver, lung, kidney, ovary, skeletal muscle and the intestine. In the adult tissues, the levels of Caspase-6 were lower than in fetal tissues but remained high in the colon, stomach, lung, kidney and liver. Immunohistological analyses revealed that active Caspase-6 was abundant in goblet cells and epithelial cells sloughing off the intestinal lining of the adult colon. These results suggest that Caspase-6 is likely important in most tissues during early development but is less involved in adult tissues. The low levels of Caspase-6 in fetal and adult brain indicate that increased expression as observed in Alzheimer Disease is a pathological condition. Lastly, the high levels of Caspase-6 in the gastrointestinal system indicate a potential specific function of Caspase-6 in these tissues.


Subject(s)
Caspase 6/metabolism , Fetus/enzymology , Gene Expression Regulation, Enzymologic , Adult , Apoptosis , Caspase 1/metabolism , Enzyme Activation , Female , Fetus/cytology , Humans , Organ Specificity , Protein Subunits/metabolism
17.
Mol Biol Cell ; 20(9): 2495-507, 2009 May.
Article in English | MEDLINE | ID: mdl-19297528

ABSTRACT

Hepatocyte growth factor/scatter factor (HGF/SF) acts through the membrane-anchored Met receptor tyrosine kinase to induce invasive growth. Deregulation of this signaling is associated with tumorigenesis and involves, in most cases, overexpression of the receptor. We demonstrate that Met is processed in epithelial cells by presenilin-dependent regulated intramembrane proteolysis (PS-RIP) independently of ligand stimulation. The proteolytic process involves sequential cleavage by metalloproteases and the gamma-secretase complex, leading to generation of labile fragments. In normal epithelial cells, although expression of cleavable Met by PS-RIP is down-regulated, uncleavable Met displayed membrane accumulation and induced ligand-independent motility and morphogenesis. Inversely, in transformed cells, the Met inhibitory antibody DN30 is able to promote Met PS-RIP, resulting in down-regulation of the receptor and inhibition of the Met-dependent invasive growth. This demonstrates the original involvement of a proteolytic process in degradation of the Met receptor implicated in negative regulation of invasive growth.


Subject(s)
Cell Membrane/enzymology , Down-Regulation , Presenilins/metabolism , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-met/metabolism , ADAM Proteins/antagonists & inhibitors , ADAM17 Protein , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Antibodies , Cell Line , Cell Membrane/drug effects , Cell Proliferation/drug effects , Dogs , Down-Regulation/drug effects , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Hepatocyte Growth Factor/pharmacology , Humans , Ligands , Metalloproteases/antagonists & inhibitors , Mice , Peptide Fragments/metabolism , Proteasome Inhibitors , Protein Processing, Post-Translational/drug effects , Protein Stability/drug effects , Recombinant Proteins/metabolism
18.
Cell Signal ; 21(9): 1455-63, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19450681

ABSTRACT

The MET tyrosine kinase is the hepatocyte growth factor/scatter factor (HGF/SF) receptor, which elicits multiple biological responses in epithelial cells, including cell survival. We previously demonstrated that in stress conditions, the MET receptor is cleaved by caspases within its juxtamembrane region, generating a pro-apoptotic intracellular fragment of 40 kDa. The caspase cleavage site at aspartic acid D1000 is adjacent to tyrosine Y1001, which when phosphorylated upon MET activation, is involved in CBL recruitment, allowing receptor ubiquitination and down regulation. Scanning mutagenesis of the MET juxtamembrane region led us to demonstrate that V999 and D1000 are essential for the caspase cleavage, while D1000 and Y1001 are essential for CBL recruitment. By examining whether overlapping of these sites leads to a functional interference, an inverse relationship was found between generation of p40 MET and phosphorylation of MET, with a direct involvement of phosphorylated Y1001 in protecting MET against its caspase cleavage. A molecular modeling analysis of caspase 3 interaction with the juxtamembrane region of MET confirmed that phosphorylation of this tyrosine is not compatible with its recognition by active caspase 3. These data demonstrate a direct protection mechanism of an activated phosphorylated MET receptor, against its caspase-dependent cleavage.


Subject(s)
Caspases/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Growth Factor/metabolism , Tyrosine/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Membrane/physiology , Computer Simulation , Dogs , HeLa Cells , Humans , Phosphorylation , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins c-cbl/metabolism , Proto-Oncogene Proteins c-met , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL