Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Gene Ther ; 31(5-6): 234-241, 2024 05.
Article in English | MEDLINE | ID: mdl-38135787

ABSTRACT

EPM1 is the most common form of Progressive Myoclonus Epilepsy characterized by late-childhood onset, ever-worsening and disabling myoclonus, seizures, ataxia, psychiatric disease, and shortened lifespan. EPM1 is caused by expansions of a dodecamer repeat sequence in the promoter of CSTB (cystatin B), which dramatically reduces, but does not eliminate, gene expression. The relatively late onset and consistent presence of a minimal amount of protein product makes EPM1 a favorable target for gene replacement therapy. If treated early, these children's normally developed brains could be rescued from the neurodegeneration that otherwise follows, and their cross-reactive immunological material (CRIM) positive status greatly reduces transgene related toxicity. We performed a proof-of-concept CSTB gene replacement study in Cstb knockout mice by introducing full-length human CSTB driven by the CBh promoter packaged in AAV9 and administered at postnatal days 21 and 60. Mice were sacrificed at 2 or 9 months of age, respectively. We observed significant improvements in expression levels of neuroinflammatory pathway genes and cerebellar granule cell layer apoptosis, as well as amelioration of motor impairment. The data suggest that gene replacement is a promising therapeutic modality for EPM1 and could spare affected children and families the ravages of this otherwise severe neurodegenerative disease.


Subject(s)
Cystatin B , Genetic Therapy , Mice, Knockout , Neuroinflammatory Diseases , Animals , Mice , Genetic Therapy/methods , Cystatin B/genetics , Neuroinflammatory Diseases/therapy , Neuroinflammatory Diseases/genetics , Humans , Ataxia/genetics , Ataxia/therapy , Myoclonic Epilepsies, Progressive/genetics , Myoclonic Epilepsies, Progressive/therapy , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors/genetics , Genetic Vectors/administration & dosage
2.
Fish Shellfish Immunol ; 124: 442-453, 2022 May.
Article in English | MEDLINE | ID: mdl-35460877

ABSTRACT

Cystatins are a diverse group of cysteine protease inhibitors widely present among various organisms. Beyond their protease inhibitor function, cystatins play a crucial role in diverse pathophysiological conditions in animals, including neurodegenerative disorders, tumor progression, inflammatory diseases, and immune response. However, the role of cystatins in immunity against viral and bacterial infections in fish remains to be elucidated. In this study, the cystatin B from big-belly seahorse, Hippocampus abdominalis, designated as HaCSTB, was identified and characterized. HaCSTB shared the highest homology with type 1 cystatin family members of teleosts and had three cystatin catalytic domains with no signal peptides or disulfide bonds. HaCSTB transcripts were mainly expressed in peripheral blood cells (PBCs), followed by the testis and pouch of healthy big-belly seahorses. Immune challenge with lipopolysaccharides (LPS), polyinosinic:polycytidylic acid (Poly I:C), and Streptococcus iniae induced upregulation of relative HaCSTB mRNA expression in PBCs. Subcellular localization analysis revealed the distribution of HaCSTB in the cytosol, mitochondria, and nuclei of fathead minnow cells (FHM). Recombinant HaCSTB (rHaCSTB) exhibited potent in vitro inhibitory activity against papain, a cysteine protease, in a concentration-, pH-, and temperature-dependent manner. Overexpression of HaCSTB in viral hemorrhagic septicemia virus (VHSV)-susceptible FHM cells increased cell viability and reduced VHSV-induced apoptosis. Collectively, these results suggest that HaCSTB might engage in the teleostean immune protection against bacteria and viruses.


Subject(s)
Cyprinidae , Cystatins , Fish Diseases , Smegmamorpha , Animals , Cyprinidae/genetics , Cystatin B/genetics , Cystatins/genetics , Fish Proteins/chemistry , Male , Phylogeny , Poly I-C/pharmacology , Sequence Alignment
3.
J Neurochem ; 158(3): 798-806, 2021 08.
Article in English | MEDLINE | ID: mdl-33675537

ABSTRACT

No robust biomarkers have yet been identified for autism spectrum disorder (ASD) or autistic traits. Familial factors likely influence biomarkers such as protein concentrations. Comparing twins with ASD or high autistic traits to the less affected co-twin allows estimating the impact of familial confounding. We measured 203 proteins in cerebrospinal fluid (n = 86) and serum (n = 127) in twins (mean age 14.2 years, 44.9% females) enriched for ASD and other neurodevelopmental conditions. Autistic traits were assessed by using the parent-report version of the Social Responsiveness Scale-2. In cerebrospinal fluid, autistic traits correlated negatively with three proteins and positively with one. In serum, autistic traits correlated positively with 15 and negatively with one. Also in serum, six were positively-and one negatively-associated with ASD. A pathway analysis of these proteins revealed immune system enrichment. In within twin pair analyses, autistic traits were associated with serum B-cell activating factor (BAFF) only, whereas Cystatin B (CSTB) remained significantly associated with ASD. These associations did not remain significant when only considering monozygotic twins. For the remainder, the within-pair analysis indicated familial confounding, including shared environment and genes, influencing both autism and protein levels. Our findings indicate proteins involved in immunity as putative biomarkers of autistic traits and ASD with partial genetic confounding. Although some results are in line with previous studies in general, further studies are needed for replication.


Subject(s)
Autism Spectrum Disorder/blood , Autism Spectrum Disorder/cerebrospinal fluid , Twins, Monozygotic , Adolescent , Adult , Autism Spectrum Disorder/diagnosis , Autism Spectrum Disorder/genetics , Autistic Disorder/blood , Autistic Disorder/cerebrospinal fluid , Autistic Disorder/diagnosis , Autistic Disorder/genetics , B-Cell Activating Factor/blood , B-Cell Activating Factor/cerebrospinal fluid , B-Cell Activating Factor/genetics , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Child , Cohort Studies , Cross-Sectional Studies , Cystatin B/blood , Cystatin B/cerebrospinal fluid , Cystatin B/genetics , Female , Humans , Male , Protein Interaction Maps/physiology , Twins, Monozygotic/genetics , Young Adult
4.
Neurobiol Dis ; 156: 105418, 2021 08.
Article in English | MEDLINE | ID: mdl-34102276

ABSTRACT

Cystatin B (CSTB) acts as an inhibitor of cysteine proteases of the cathepsin family and loss-of-function mutations result in human brain diseases with a genotype-phenotype correlation. In the most severe case, CSTB-deficiency disrupts brain development, and yet the molecular basis of this mechanism is missing. Here, we establish CSTB as a regulator of chromatin structure during neural stem cell renewal and differentiation. Murine neural precursor cells (NPCs) undergo transient proteolytic cleavage of the N-terminal histone H3 tail by cathepsins B and L upon induction of differentiation into neurons and glia. In contrast, CSTB-deficiency triggers premature H3 tail cleavage in undifferentiated self-renewing NPCs and sustained H3 tail proteolysis in differentiating neural cells. This leads to significant transcriptional changes in NPCs, particularly of nuclear-encoded mitochondrial genes. In turn, these transcriptional alterations impair the enhanced mitochondrial respiration that is induced upon neural stem cell differentiation. Collectively, our findings reveal the basis of epigenetic regulation in the molecular pathogenesis of CSTB deficiency.


Subject(s)
Cystatin B/deficiency , Histones/metabolism , Neural Stem Cells/metabolism , Neurogenesis/physiology , Animals , Cells, Cultured , Cystatin B/genetics , Epigenesis, Genetic/physiology , Histones/genetics , Mice , Mice, 129 Strain , Mice, Knockout
5.
Epilepsia ; 61(11): 2593-2608, 2020 11.
Article in English | MEDLINE | ID: mdl-32940364

ABSTRACT

OBJECTIVE: Microglial phagocytosis of apoptotic cells is an essential component of the brain regenerative response during neurodegeneration. Whereas it is very efficient in physiological conditions, it is impaired in mouse and human mesial temporal lobe epilepsy, and now we extend our studies to a model of progressive myoclonus epilepsy type 1 in mice lacking cystatin B (CSTB). METHODS: We used confocal imaging and stereology-based quantification of apoptosis and phagocytosis of the hippocampus of Cstb knockout (KO) mice, an in vitro model of phagocytosis and siRNAs to acutely reduce Cstb expression, and a virtual three-dimensional (3D) model to analyze the physical relationship between apoptosis, phagocytosis, and active hippocampal neurons. RESULTS: Microglial phagocytosis was impaired in the hippocampus of Cstb KO mice at 1 month of age, when seizures arise and hippocampal atrophy begins. This impairment was not related to the lack of Cstb in microglia alone, as shown by in vitro experiments with microglial Cstb depletion. The phagocytosis impairment was also unrelated to seizures, as it was also present in Cstb KO mice at postnatal day 14, before seizures begin. Importantly, phagocytosis impairment was restricted to the granule cell layer and spared the subgranular zone, where there are no active neurons. Furthermore, apoptotic cells (both phagocytosed and not phagocytosed) in Cstb-deficient mice were at close proximity to active cFos+ neurons, and a virtual 3D model demonstrated that the physical relationship between apoptotic cells and cFos+ neurons was specific for Cstb KO mice. SIGNIFICANCE: These results suggest a complex crosstalk between apoptosis, phagocytosis, and neuronal activity, hinting that local neuronal activity could be related to phagocytosis dysfunction in Cstb KO mice. Overall, these data suggest that phagocytosis impairment is an early feature of hippocampal damage in epilepsy and opens novel therapeutic approaches for epileptic patients based on targeting microglial phagocytosis.


Subject(s)
Dentate Gyrus/metabolism , Disease Models, Animal , Microglia/metabolism , Neurons/metabolism , Phagocytosis/physiology , Unverricht-Lundborg Syndrome/metabolism , Animals , Cystatin B/deficiency , Cystatin B/genetics , Dentate Gyrus/pathology , Mice , Mice, Knockout , Microglia/pathology , Neurons/pathology , Unverricht-Lundborg Syndrome/genetics , Unverricht-Lundborg Syndrome/pathology
6.
Fish Shellfish Immunol ; 105: 78-85, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32645518

ABSTRACT

Cystatins B is an endogenous cysteine cathepsin inhibitor. In shrimp, cystatins B-like (CSTB-L) has not been characterized and its role in WSSV infection is largely unknown. In this study, a full-length 699 bp CSTB-L sequence with 291 bp open reading frame encoding a 96 amino acid from L.vannamei (Lv) was first cloned. The tissue distribution assay indicated that LvCSTB-L presented ubiquitous expression in most examined tissues, with the most predominant expression in the hepatopancreas and the weakest expression in the muscles. LvCSTB-L transcripts could be induced in the intestine and hepatopancreas by WSSV challenge. The relative expression level of IE1 and VP28 in the LvCSTB-L knockdown shrimp were increased significantly. In addition, the shrimp cumulative mortality was remarkably (p < 0.01) increased after LvCSTB-L knockdown. Moreover, following the LvCSTB-L silencing, significant decreases in the mRNA levels of p53, p38, caspase3, STAT and ERK were also observed. The results suggested that LvCSTB-L could play positively roles in antiviral immune response by JAK-STAT, MAPK and apoptotic pathway. These findings would further our understanding of shrimp antiviral response, and therefore help for virus control and prevention.


Subject(s)
Cystatin B/genetics , Cystatin B/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Penaeidae/genetics , Penaeidae/immunology , Amino Acid Sequence , Animals , Arthropod Proteins/chemistry , Arthropod Proteins/genetics , Arthropod Proteins/immunology , Base Sequence , Cystatin B/chemistry , Gene Expression Profiling , Phylogeny , Sequence Alignment
7.
Fish Shellfish Immunol ; 104: 497-505, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32534230

ABSTRACT

Cystatins represent a large superfamily of proteins involved in the competitive reversible inhibition of C1 class cysteine proteases. Plant-derived papain proteases and cysteine cathepsins are the major cysteine proteases that interact with cystatins. The cystatin superfamily can be further classified into three groups: stefins, cystatins, and kininogens. Among these, cystatin B is categorized under stefins. Cystatin B lacks a signal sequence, disulfide bonds, and carbohydrate groups. However, it contains the conserved cystatin family signature, including a single cystatin-like domain, cysteine protease inhibitory signature concealing pentapeptide (QXVXG) consensus sequence, and two conserved neighboring glycine (8GG9) residues at the N-terminal. In the current study, a member of cystatin B was identified from Korean black rockfish (Sebastes schlegeli) using a cDNA database and designated as RfCytB. The full-length cDNA of RfCytB was 573 bp long, with a coding region of 294 bp. The 5'-untranslated region (UTR) comprised 55 bp, and the 263-bp-long 3'-UTR included a polyadenylation signal sequence and a poly-A tail. The coding sequence encodes a polypeptide comprising 97 amino acids, with a predicted molecular weight of 11 kDa and theoretical isoelectric point of 6.3. RfCytB shared homology features with similar molecules from other teleost and vertebrate species, and was clustered with Cystatin family 1 in our phylogenetic reconstruction. RfCytB was ubiquitously expressed in all tissue types of healthy animals, with the highest levels of expression observed in gill and spleen. Temporal expression of RfCytB displayed significant up-regulation upon infection with Aeromonas salmonicida. Recombinantly expressed RfCytB showed a concentration-dependent inhibitory activity towards papain, with a high thermal stability. Transient expression of RfCytB in LPS activated murine macrophages, thereby inducing the expression of genes related to pro-inflammatory conditions, such as iNOS and TNF α. These results provide evidence for its protease inhibitory and immunity relevant roles in hosts.


Subject(s)
Cystatin B/genetics , Cystatin B/immunology , Fish Diseases/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Perciformes/genetics , Perciformes/immunology , Amino Acid Sequence , Animals , Cystatin B/chemistry , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Fishes , Gene Expression Profiling/veterinary , Phylogeny , Sequence Alignment/veterinary
8.
Epilepsy Behav ; 112: 107439, 2020 11.
Article in English | MEDLINE | ID: mdl-32920378

ABSTRACT

AIM OF THE STUDY: The aim of this study was to explore genetic findings and the phenotype in Polish patients with Unverricht-Lundborg disease (ULD). MATERIALS AND METHODS: We retrospectively evaluated mutations in the cystatin B (CSTB) gene and clinical presentation in a cohort of patients with ULD. The study population consisted of 19 (14 males) patients with genetically confirmed disease. RESULTS: Sixteen patients were homozygous for the expanded dodecamer repeat mutation alleles, one subject was compound heterozygous for the dodecamer repeat expansion and other mutation, in two, the type of mutation has not yet been established. The numbers of repeats in the CSTB gene varied from 60 to 81. Clinical information was available for 16 subjects. The disease course was progressive in all patients, leading to severe disability, mainly due to myoclonus, in nine. CONCLUSIONS AND CLINICAL IMPLICATIONS: Genetic findings and the clinical picture of our patients with ULD were in accordance with available studies. The most common genetic defect underlying ULD was homozygosity for an unstable expansion of a dodecamer repeat in the CSTB gene. Patients with action or/and stimulus sensitive myoclonus or intractable myoclonus epilepsy, especially with onset in late childhood/adolescence should be screened for ULD.


Subject(s)
Unverricht-Lundborg Syndrome , Adolescent , Child , Cohort Studies , Cystatin B/genetics , Genetic Testing , Humans , Male , Phenotype , Poland , Retrospective Studies , Unverricht-Lundborg Syndrome/genetics
9.
J Cell Biochem ; 120(6): 10662-10669, 2019 06.
Article in English | MEDLINE | ID: mdl-30652348

ABSTRACT

Earlier studies showed that the oxidant menadione (MD) induces apoptosis in certain cells and also has anticancer effects. Most of these studies emphasized the role of the mitochondria in this process. However, the engagement of other organelles is less known. Particularly, the role of lysosomes and their proteolytic system, which participates in apoptotic cell death, is still unclear. The aim of this study was to investigate the role of lysosomal cathepsins on molecular signaling in MD-induced apoptosis in U937 cells. MD treatment induced translocation of cysteine cathepsins B, C, and S, and aspartic cathepsin D. Once in the cytosol, some cathepsins cleaved the proapoptotic molecule, Bid, in a process that was completely prevented by E64d, a general inhibitor of cysteine cathepsins, and partially prevented by the pancaspase inhibitor, z-VAD-fmk. Upon loss of the mitochondrial membrane potential, apoptosome activation led to caspase-9 processing, activation of caspase-3-like caspases, and poly (ADP-ribose) polymerase cleavage. Notably, the endogenous protein inhibitor, stefin B, was degraded by cathepsin D and caspases. This process was prevented by z-VAD-fmk, and partially by pepstatin A-penetratin. These findings suggest that the cleaved Bid protein acts as an amplifier of apoptotic signaling through mitochondria, thus enhancing the activity of cysteine cathepsins following stefin B degradation.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/genetics , Cystatin B/genetics , Gene Expression Regulation, Neoplastic , Lysosomes/drug effects , Vitamin K 3/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis/genetics , Apoptosomes/drug effects , Apoptosomes/metabolism , BH3 Interacting Domain Death Agonist Protein/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cathepsin B/antagonists & inhibitors , Cathepsin B/genetics , Cathepsin B/metabolism , Cathepsin C/antagonists & inhibitors , Cathepsin C/genetics , Cathepsin C/metabolism , Cathepsin D/antagonists & inhibitors , Cathepsin D/genetics , Cathepsin D/metabolism , Cathepsins/antagonists & inhibitors , Cathepsins/genetics , Cathepsins/metabolism , Cystatin B/metabolism , Humans , Leucine/analogs & derivatives , Leucine/pharmacology , Lysosomes/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Pepstatins/pharmacology , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Protease Inhibitors/pharmacology , Proteolysis/drug effects , Signal Transduction , U937 Cells
10.
Int J Med Sci ; 16(10): 1338-1348, 2019.
Article in English | MEDLINE | ID: mdl-31692912

ABSTRACT

Endometrial cancer is one of the most common cancers in women worldwide, affecting more than 300,000 women annually. Dysregulated gene expression, especially those mediated by microRNAs, play important role in the development and progression of cancer. This study aimed to investigate differentially expressed genes in endometrial adenocarcinoma using next generation sequencing (NGS) and bioinformatics. The gene expression profiles and microRNA profiles of endometrial adenocarcinoma (cancer part) and normal endometrial tissue (non-cancer part) were assessed with NGS. We identified 56 significantly dysregulated genes, including 47 upregulated and 9 downregulated genes, in endometrial adenocarcinoma. Most of these genes were associated with defense response, response to stimulus, and immune system process, and further pathway analysis showed that human papillomavirus infection was the most significant pathway in endometrial adenocarcinoma. In addition, these genes were also associated with decreased cell death and survival as well as increased cellular movement. The analyses using Human Protein Atlas, identified 6 genes (PEG10, CLDN1, ASS1, WNT7A, GLDC, and RSAD2) significantly associated with poorer prognosis and 3 genes (SFN, PIGR, and CDKN1A) significantly associated with better prognosis. Combining with the data of microRNA profiles using microRNA target predicting tools, two significantly dysregulated microRNA-mediated gene expression changes in endometrial adenocarcinoma were identified: downregulated hsa-miR-127-5p with upregulated CSTB and upregulated hsa-miR-218-5p with downregulated HPGD. These findings may contribute important new insights into possible novel diagnostic or therapeutic strategies for endometrial adenocarcinoma.


Subject(s)
Adenocarcinoma/genetics , Biomarkers, Tumor/metabolism , Endometrial Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Adenocarcinoma/immunology , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Aged , Biomarkers, Tumor/genetics , Computational Biology , Cystatin B/genetics , Down-Regulation , Endometrial Neoplasms/immunology , Endometrial Neoplasms/mortality , Endometrial Neoplasms/pathology , Endometrium/pathology , Female , Follow-Up Studies , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Hydroxyprostaglandin Dehydrogenases/genetics , Kaplan-Meier Estimate , MicroRNAs/metabolism , Middle Aged , Prognosis , Signal Transduction/genetics , Signal Transduction/immunology , Up-Regulation
11.
Hum Mol Genet ; 25(7): 1434-46, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26908626

ABSTRACT

Niemann-Pick C1 (NPC) disease, an autosomal recessive lipid trafficking disorder caused by loss-of-function mutations in the NPC1 gene, is characterized by progressive neurodegeneration resulting in cognitive impairment, ataxia and early death. Little is known about the cellular pathways leading to neuron loss. Here, we studied the effects of diminishing expression of cystatin B, an endogenous inhibitor of cathepsins B, H and L, on the development of NPC neuropathology. We show that decreased expression of cystatin B in patient fibroblasts enhances cathepsin activity. Deletion of the encoding Cstb gene in Npc1-deficient mice resulted in striking deleterious effects, particularly within the cerebellum where diffuse loss of Purkinje cells was observed in young mice. This severe pathology occurred through cell autonomous mechanisms that triggered Purkinje cell death. Moreover, our analyses demonstrated the mislocalization of lysosomal cathepsins within the cytosol of Npc1-deficient Purkinje cells. We provide evidence that this may be a consequence of damage to lysosomal membranes by reactive oxygen species (ROS), leading to the leakage of lysosomal contents that culminates in apoptotic cell death. Consistent with this notion, toxicity from ROS was attenuated in an NPC cell model by cystatin B over-expression or pharmacological inhibition of cathepsin B. The observation that Npc1 and Cstb deletion genetically interact to potently enhance the degenerative phenotype of the NPC cerebellum provides strong support for the notion that lysosomal membrane permeabilization contributes to cerebellar degeneration in NPC disease.


Subject(s)
Cathepsin B/metabolism , Cystatin B/metabolism , Nerve Degeneration , Niemann-Pick Disease, Type C/metabolism , Purkinje Cells/metabolism , Animals , Apoptosis , Carrier Proteins/genetics , Cathepsin B/antagonists & inhibitors , Cystatin B/genetics , Cystatin B/pharmacology , Down-Regulation , Humans , Intracellular Signaling Peptides and Proteins , Lysosomes/metabolism , Lysosomes/pathology , Membrane Glycoproteins/genetics , Mice , Mutation , Niemann-Pick C1 Protein , Niemann-Pick Disease, Type C/genetics , Niemann-Pick Disease, Type C/pathology , Oxidative Stress , Proteins/genetics , Purkinje Cells/pathology
12.
Fish Shellfish Immunol ; 81: 463-469, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30064019

ABSTRACT

Cystatin B is an intracellular inhibitor that regulates the activities of cysteine proteases. In this study, cystatin B in Japanese flounder (Paralichthys olivaceus) was characterized and its immune function was analyzed. This gene had a high similarity with the sequence of cystatin B in other fish species, and the derived peptide shared typical features of cystatin proteins including the QXVXG motif. The results of quantitative real-time PCR showed that cystatin B mRNA was constitutively expressed in all examined tissues, with the highest level in gill. The stimulations of lipopolysaccharide, peptidoglycan and polyinosinic-polycytidylic acid effectively increased the expression level of cystatin B mRNA. Functional analysis implied that the recombinant P. olivaceus cystatin B purified from Escherichia coli had cysteine protease inhibitory activity and could inhibit bacterial growth by binding to bacteria. Furthermore, we found that P. olivaceus cystatin B had no effects on the expression of inflammatory factors cytokines tumor necrosis factor α, interleukin 10, interleukin 1ß and interferon γ. These results indicate that cystatin B of P. olivaceus is potentially involved in immune responses against invading microbial pathogens, and provide a better understanding of the immune mechanisms of cystatins in teleosts.


Subject(s)
Cystatin B/immunology , Flounder/immunology , Animals , Cell Line , Cystatin B/genetics , Cystatin B/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Cytokines/genetics , DNA, Complementary/genetics , Female , Flounder/genetics , Immunologic Factors/pharmacology , Lipopolysaccharides/pharmacology , Male , Peptidoglycan/pharmacology , Poly I-C/pharmacology , Recombinant Proteins/pharmacology
13.
J Mol Recognit ; 30(1)2017 01.
Article in English | MEDLINE | ID: mdl-27577977

ABSTRACT

We describe studies performed thus far on stefin B from the family of cystatins as a model protein for folding and amyloid fibril formation studies. We also briefly mention our studies on aggregation of some of the missense EPM1 mutants of stefin B in cells, which mimic additional pathological traits (gain in toxic function) in selected patients with EPM1 disease. We collected data on the reported interactors of stefin B and discuss several hypotheses of possible cytosolic alternative functions.


Subject(s)
Amyloid beta-Peptides/metabolism , Cell Membrane/metabolism , Copper/metabolism , Cystatin B/chemistry , Cystatin B/metabolism , Binding Sites , Cystatin B/genetics , Humans , Models, Molecular , Mutation, Missense , Protein Binding , Protein Conformation , Protein Folding
14.
Epilepsia ; 58(2): e31-e35, 2017 02.
Article in English | MEDLINE | ID: mdl-27888502

ABSTRACT

Unverricht-Lundborg disease or progressive myoclonic epilepsy type 1 (EPM1) is an autosomal recessive disease caused by mutation of the cystatin B gene (CSTB), located on chromosome 21q22.3. The most common mutation is an expansion of unstable dodecamer repetition (CCCCGCCCCGCG), whereas other types of mutations are rare. Among these, heterozygous compound mutations are described to induce a more severe phenotype than that of homozygous dodecameric repetition. We report two siblings affected by heterozygous compound mutations carrying a novel mutation of the deletion of three nucleotides in exon 2 of the gene in position 132-134 of the coding sequence (c.132-134del) in the allele not including the dodecamer repetition. This mutation results in the loss of two amino acid residues and insertion of an asparagine in position 44 (p.Lys44_Ser45delinsAsn). Our patients presented a very different clinical picture. The male patient had a severe myoclonus, drug-resistant epilepsy and psychiatric comorbidity, while his affected sister had only very rare seizures and sporadic myoclonic jerks at awakening. The revision of literature about heterozygous compound EPM1 patients confirms this gender phenotypic expressivity, with female patients carrying less severe symptoms than male patients. These data lead to the hypothesis of complex gender-specific factors interacting with CSTB expressivity in EPM1 patients.


Subject(s)
Cystatin B/genetics , Sequence Deletion/genetics , Unverricht-Lundborg Syndrome/genetics , Chromosomes, Human, Pair 21/genetics , DNA Mutational Analysis , Female , Humans , Male , Middle Aged , Phenotype , Siblings
15.
Mol Cell Proteomics ; 14(11): 3000-14, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26269332

ABSTRACT

The tau protein is central to the etiology of several neurodegenerative diseases, including Alzheimer's disease, a subset of frontotemporal dementias, progressive supranuclear palsy and dementia following traumatic brain injury, yet the proteins it interacts with have not been studied using a systematic discovery approach. Here we employed mild in vivo crosslinking, isobaric labeling, and tandem mass spectrometry to characterize molecular interactions of human tau in a neuroblastoma cell model. The study revealed a robust association of tau with the ribonucleoproteome, including major protein complexes involved in RNA processing and translation, and documented binding of tau to several heat shock proteins, the proteasome and microtubule-associated proteins. Follow-up experiments determined the relative contribution of cellular RNA to the tau interactome and mapped interactions to N- or C-terminal tau domains. We further document that expression of P301L mutant tau disrupts interactions of the C-terminal half of tau with heat shock proteins and the proteasome. The data are consistent with a model whereby a higher propensity of P301L mutant tau to aggregate may reflect a perturbation of its chaperone-assisted stabilization and proteasome-dependent degradation. Finally, using a global proteomics approach, we show that heterologous expression of a tau construct that lacks the C-terminal domain, including the microtubule binding domain, does not cause a discernible shift of the proteome except for a significant direct correlation of steady-state levels of tau and cystatin B.


Subject(s)
Epithelial Cells/metabolism , Molecular Chaperones/metabolism , Neurons/metabolism , Proteasome Endopeptidase Complex/metabolism , Ribonucleoproteins/metabolism , tau Proteins/metabolism , Animals , Binding Sites , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Cystatin B/genetics , Cystatin B/metabolism , Epithelial Cells/cytology , Gene Expression Regulation , HEK293 Cells , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Molecular Chaperones/genetics , Molecular Sequence Annotation , Mutation , Neurons/cytology , Protein Binding , Protein Interaction Mapping , Protein Structure, Tertiary , Ribonucleoproteins/genetics , Signal Transduction , tau Proteins/genetics
16.
J Neuroinflammation ; 13(1): 298, 2016 11 28.
Article in English | MEDLINE | ID: mdl-27894304

ABSTRACT

Progressive myoclonus epilepsy of Unverricht-Lundborg type (EPM1) is an autosomal recessively inherited childhood-onset neurodegenerative disorder, characterized by myoclonus, seizures, and ataxia. Mutations in the cystatin B gene (CSTB) underlie EPM1. The CSTB-deficient (Cstb -/- ) mouse model recapitulates key features of EPM1, including myoclonic seizures. The mice show early microglial activation that precedes seizure onset and neuronal loss and leads to neuroinflammation. We here characterized the inflammatory phenotype of Cstb -/- mice in more detail. We found higher concentrations of chemokines and pro-inflammatory cytokines in the serum of Cstb -/- mice and higher CXCL13 expression in activated microglia in Cstb -/- compared to control mouse brains. The elevated chemokine levels were not accompanied by blood-brain barrier disruption, despite increased brain vascularization. Macrophages in the spleen and brain of Cstb -/- mice were predominantly pro-inflammatory. Taken together, these data show that CXCL13 expression is a hallmark of microglial activation in Cstb -/- mice and that the brain inflammation is linked to peripheral inflammatory changes, which might contribute to the disease pathology of EPM1.


Subject(s)
Cystatin B/deficiency , Encephalitis/etiology , Gene Expression Regulation/genetics , Inflammation/etiology , Myoclonic Epilepsies, Progressive/complications , Myoclonic Epilepsies, Progressive/genetics , Animals , Brain/pathology , Cystatin B/genetics , Cytokines/blood , Disease Models, Animal , Encephalitis/blood , Inflammation/blood , Mice , Mice, Knockout , Microglia/metabolism
17.
J Neurovirol ; 22(5): 666-673, 2016 10.
Article in English | MEDLINE | ID: mdl-27137788

ABSTRACT

Cystatin B is a cysteine protease inhibitor that induces HIV replication in monocyte-derived macrophages (MDM). This protein interacts with signal transducer and activator of transcription (STAT-1) factor and inhibits the interferon (IFN-ß) response in Vero cells by preventing STAT-1 translocation to the nucleus. Cystatin B also decreases the levels of tyrosine-phosphorylated STAT-1 (STAT-1PY). However, the mechanisms of cystatin B regulation on STAT-1 phosphorylation in MDM are unknown. We hypothesized that cystatin B inhibits IFN-ß antiviral responses and induces HIV replication in macrophage reservoirs through the inhibition of STAT-1 phosphorylation. Macrophages were transfected with cystatin B siRNA prior to interferon-ß treatment or infected with HIV-ADA to determine the effect of cystatin B modulation in STAT-1 localization and activation using immunofluorescence and proximity ligation assays. Cystatin B decreased STAT-1PY and its transportation to the nucleus, while HIV infection retained unphosphorylated STAT (USTAT-1) in the nucleus avoiding its exit to the cytoplasm for eventual phosphorylation. In IFN-ß-treated MDM, cystatin B inhibited the nuclear translocation of both, USTAT-1 and STAT-1PY. These results demonstrate that cystatin B interferes with the STAT-1 signaling and IFN-ß-antiviral responses perpetuating HIV in macrophage reservoirs.


Subject(s)
Cystatin B/genetics , HIV-1/immunology , Host-Pathogen Interactions , Interferon-beta/pharmacology , Macrophages/drug effects , STAT1 Transcription Factor/genetics , Cell Nucleus/drug effects , Cell Nucleus/immunology , Cell Nucleus/virology , Cystatin B/antagonists & inhibitors , Cystatin B/immunology , Gene Expression Regulation , HIV-1/growth & development , Humans , Macrophages/immunology , Macrophages/virology , Phosphorylation/drug effects , Primary Cell Culture , Protein Transport/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , STAT1 Transcription Factor/immunology , Signal Transduction , Transfection , Virus Replication/drug effects
18.
Protein Expr Purif ; 118: 10-7, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26481272

ABSTRACT

Cystatins are reversible cysteine protease inhibitor proteins. They are known to play important roles in controlling cathepsins, neurodegenerative disease, and in immune system regulation. Production of recombinant cystatin proteins is important for biochemical and function characterization. In this study, we cloned and expressed human stefin A, stefin B and cystatin C in Escherichia coli. Human stefin A, stefin B and cystatin C were purified from soluble fraction. For cystatin C, we used various chaperone plasmids to make cystatin C soluble, as it is reported to localize in inclusion bodies. Trigger factor, GroES-GroEL, DnaK-DnaJ-GrpE chaperones lead to the presence of cystatin C in the soluble fraction. Immobilized metal affinity chromatography, glutathione sepharose and anion exchange chromatography techniques were employed for efficient purification of these proteins. Their biological activities were tested by inhibition assays against cathepsin L and H3 protease.


Subject(s)
Cystatin A/genetics , Cystatin A/isolation & purification , Cystatin B/genetics , Cystatin B/isolation & purification , Cystatin C/genetics , Cystatin C/isolation & purification , Cathepsin L/antagonists & inhibitors , Cathepsin L/chemistry , Cystatin A/chemistry , Cystatin A/metabolism , Cystatin B/chemistry , Cystatin B/metabolism , Cystatin C/chemistry , Cystatin C/metabolism , Endopeptidases/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/isolation & purification , Enzyme Inhibitors/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Kinetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
19.
Biochim Biophys Acta ; 1843(9): 2089-99, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24909779

ABSTRACT

EPM1 is a rare progressive myoclonus epilepsy accompanied by apoptosis in the cerebellum of patients. Mutations in the gene of stefin B (cystatin B) are responsible for the primary defect underlying EPM1. Taking stefin B aggregates as a model we asked what comes first, protein aggregation or oxidative stress, and how these two processes correlate with cell death. We studied the aggregation in cells of the stefin B wild type, G4R mutant, and R68X fragment before (Ceru et al., 2010, Biol. Cell). The present study was performed on two more missense mutants of human stefin B, G50E and Q71P, and they similarly showed numerous aggregates upon overexpression. Mutant- and oligomer-dependent increase in oxidative stress and cell death in cells bearing aggregates was shown. On the other hand, there was no correlation between the size and number of the aggregates and cell death. We suggest that differences in toxicity of the aggregates depend on whether they are in oligomeric/protofibrillar or fibrillar form. This in turn likely depends on the mutant's 3D structure where unfolded proteins show lower toxicity. Imaging by transmission electron microscopy showed that the aggregates in cells are of different types: bigger perinuclear, surrounded by membranes and sometimes showing vesicle-like invaginations, or smaller, punctual and dispersed throughout the cytoplasm. All EPM1 mutants studied were inactive as cysteine proteases inhibitors and in this way contribute to loss of stefin B functions. Relevance to EPM1 disease by gain in toxic function is discussed.


Subject(s)
Cystatin B/chemistry , Cystatin B/genetics , Mutant Proteins/chemistry , Oxidative Stress , Unverricht-Lundborg Syndrome/genetics , Unverricht-Lundborg Syndrome/pathology , Amyloid/metabolism , Animals , Annexin A5/metabolism , Benzothiazoles , CHO Cells , Cell Count , Cell Death , Cell Survival , Cricetinae , Cricetulus , Cystatin B/ultrastructure , HEK293 Cells , Humans , Kinetics , Mutant Proteins/ultrastructure , Propidium/metabolism , Protein Structure, Quaternary , Spectrometry, Fluorescence , Thiazoles/metabolism , Transfection
20.
Glia ; 63(3): 400-11, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25327891

ABSTRACT

Progressive myoclonus epilepsy of Unverricht-Lundborg type (EPM1) is an autosomal-recessively inherited neurodegenerative disorder characterized by severely incapacitating myoclonus, seizures, and ataxia, and caused by loss-of-function mutations in the cystatin B gene (CSTB). A central neuropathological finding in the Cstb(-/-) mouse, an animal model for EPM1, is early microglial activation, which precedes astroglial activation, neuronal loss, and onset of myoclonus, thus implying a critical role for microglia in EPM1 pathogenesis. Here, we characterized phenotypic and functional properties of microglia from Cstb(-/-) mice utilizing brain tissue, microglia directly isolated from the brain, and primary microglial cultures. Our results show significantly higher Cstb mRNA expression in microglia than in neurons and astrocytes. In Cstb(-/-) mouse brain, expression of the inflammatory marker p-p38 MAPK and the proportion of both pro-inflammatory M1 and anti-inflammatory M2 microglia is higher than in control mice. Moreover, M1/M2 polarization of microglia in presymptomatic Cstb(-/-) mice is, compared to control mice, skewed towards M2 type at postnatal day 14 (P14), but towards M1 type at P30, a time point associated with onset of myoclonus. At this age, the high expression of both pro-inflammatory inducible nitric oxide synthase (iNOS) and anti-inflammatory arginase 1 (ARG1) in Cstb(-/-) mouse cortex is accompanied by the presence of peripheral immune cells. Consistently, activated Cstb(-/-) microglia show elevated chemokine release and chemotaxis. However, their MHCII surface expression is suppressed. Taken together, our results link CSTB deficiency to neuroinflammation with early activation and dysfunction of microglia and will open new avenues for therapeutic interventions for EPM1.


Subject(s)
Brain/immunology , Cystatin B/deficiency , Microglia/physiology , Unverricht-Lundborg Syndrome/immunology , Animals , Arginase/metabolism , Astrocytes/metabolism , Cells, Cultured , Cystatin B/genetics , Disease Models, Animal , Genes, MHC Class II/physiology , Granulocytes/physiology , Macrophages/physiology , Mice, 129 Strain , Neuroimmunomodulation/physiology , Neurons/metabolism , Nitric Oxide Synthase Type II/metabolism , RNA, Messenger/metabolism , T-Lymphocytes/physiology , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL