Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
Add more filters











Publication year range
1.
J Alzheimers Dis ; 101(1): 211-221, 2024.
Article in English | MEDLINE | ID: mdl-39121120

ABSTRACT

Background: Laminopathy is a pathological manifestation observed in Alzheimer's disease (AD), leading to neuronal apoptosis. Objective: Our objective was to assess inhibitors of enzymes involved in laminopathy. Methods: The mRNA expression of the cathepsins L and B, caspases 3 and 6, lamins b1 and b2, granzymes A and B, and lamins A and C were extracted and analyzed from GSE5281 and GSE28146 datasets. A total of 145 ligands were selected for molecular docking. Subsequently, 10 ns and 100 ns atomistic molecular dynamics (MD) and Martini 3 were performed with NAMD for two selected ligands (PubChem id: 608841 and ChEMBL id: 550872). Results: The mRNA expression level highlighted caspase 6 and lamin A/C upregulation in the hippocampus of the AD samples, in contrast to cathepsin B, lamin b2, and caspase 3. Moreover, there was a strong correlation between the expression level of cathepsin B, lamin A/C, and caspase 6 in the AD group. The MD results suggested molecule with ChEMBL id of 550872 had higher free binding energy, while in longer simulation the molecule with PubChem id of 608841 was suggested to be more stable in complex with the receptor. Conclusions: Our findings suggest that lamins A/C, cathepsins B/L, caspase 6, and lamin B2 are associated with laminopathy as potential factors contributing to apoptosis in AD. We propose that simultaneous inhibition of caspases 6 and cathepsins L may decrease the rate of apoptosis triggered by lamin degradation. Nevertheless, further studies are required to confirm these observations due to the lack of in vivo findings.


Subject(s)
Alzheimer Disease , Apoptosis , Caspase 3 , Caspase 6 , Cathepsin B , Cathepsin L , Humans , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Caspase 6/metabolism , Caspase 6/genetics , Apoptosis/drug effects , Cathepsin L/metabolism , Cathepsin L/genetics , Cathepsin B/metabolism , Cathepsin B/genetics , Caspase 3/metabolism , Laminopathies/genetics , Molecular Docking Simulation , Lamin Type A/genetics , Lamin Type A/metabolism , Hippocampus/pathology , Hippocampus/metabolism , Male
2.
Neurobiol Dis ; 190: 106368, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38040383

ABSTRACT

In Huntington disease, cellular toxicity is particularly caused by toxic protein fragments generated from the mutant huntingtin (HTT) protein. By modifying the HTT protein, we aim to reduce proteolytic cleavage and ameliorate the consequences of mutant HTT without lowering total HTT levels. To that end, we use an antisense oligonucleotide (AON) that targets HTT pre-mRNA and induces partial skipping of exon 12, which contains the critical caspase-6 cleavage site. Here, we show that AON-treatment can partially restore the phenotype of YAC128 mice, a mouse model expressing the full-length human HTT gene including 128 CAG-repeats. Wild-type and YAC128 mice were treated intracerebroventricularly with AON12.1, scrambled AON or vehicle starting at 6 months of age and followed up to 12 months of age, when MRI was performed and mice were sacrificed. AON12.1 treatment induced around 40% exon skip and protein modification. The phenotype on body weight and activity, but not rotarod, was restored by AON treatment. Genes differentially expressed in YAC128 striatum changed toward wild-type levels and striatal volume was preserved upon AON12.1 treatment. However, scrambled AON also showed a restorative effect on gene expression and appeared to generally increase brain volume.


Subject(s)
Huntington Disease , Animals , Humans , Mice , Caspase 6/genetics , Caspase 6/metabolism , Corpus Striatum/metabolism , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Oligonucleotides, Antisense/pharmacology , Phenotype
3.
Cell Death Differ ; 29(3): 657-669, 2022 03.
Article in English | MEDLINE | ID: mdl-34625662

ABSTRACT

The sequential activation of Nucleotide-binding oligomerization domain, Leucine rich Repeat and Pyrin domain containing protein 1 (Nlrp1) inflammasome, Caspase-1 (Casp1), and Caspase-6 (Casp6) is implicated in primary human neuron cultures and Alzheimer Disease (AD) neurodegeneration. To validate the Nlrp1-Casp1-Casp6 pathway in vivo, the APPSwedish/Indiana J20 AD transgenic mouse model was generated on either a Nlrp1, Casp1 or Casp6 null genetic background and mice were studied at 4-5 months of age. Episodic memory deficits assessed with novel object recognition were normalized by genetic ablation of Nlrp1, Casp1, or Casp6 in J20 mice. Spatial learning deficits, assessed with the Barnes Maze, were normalized in genetically ablated J20, whereas memory recall was normalized in J20/Casp1-/- and J20/Casp6-/-, and improved in J20/Nlrp1-/- mice. Hippocampal CA1 dendritic spine density of the mushroom subtype was reduced in J20, and normalized in genetically ablated J20 mice. Reduced J20 hippocampal dentate gyrus and CA3 synaptophysin levels were normalized in genetically ablated J20. Increased Iba1+-microglia in the hippocampus and cortex of J20 brains were normalized by Casp1 and Casp6 ablation and reduced by Nlrp1 ablation. Increased pro-inflammatory cytokines, TNF-α and CXCL1, in the J20 hippocampus were normalized by Nlrp1 or Casp1 genetic ablation. CXCL1 was also normalized by Casp6 genetic ablation. IFN-γ was increased and total amyloid ß peptide was decreased in genetically ablated Nlrp1, Casp1 or Casp6 J20 hippocampi. We conclude that Nlrp1, Casp1, or Casp6 are implicated in AD-related cognitive impairment, inflammation, and amyloidogenesis. These results indicate that Nlrp1, Casp1, and Casp6 represent rational therapeutic targets against cognitive impairment and inflammation in AD.


Subject(s)
Alzheimer Disease , Caspase 1/metabolism , Caspase 6/metabolism , Cognitive Dysfunction , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Caspase 6/genetics , Cognitive Dysfunction/genetics , Disease Models, Animal , Inflammasomes/metabolism , Inflammation , Mice , Mice, Transgenic
4.
Sci Rep ; 11(1): 12695, 2021 06 16.
Article in English | MEDLINE | ID: mdl-34135352

ABSTRACT

Caspase-6 (Casp6) is implicated in Alzheimer disease (AD) cognitive impairment and pathology. Hippocampal atrophy is associated with cognitive impairment in AD. Here, a rare functional exonic missense CASP6 single nucleotide polymorphism (SNP), causing the substitution of asparagine with threonine at amino acid 73 in Casp6 (Casp6N73T), was associated with hippocampal subfield CA1 volume preservation. Compared to wild type Casp6 (Casp6WT), recombinant Casp6N73T altered Casp6 proteolysis of natural substrates Lamin A/C and α-Tubulin, but did not alter cleavage of the Ac-VEID-AFC Casp6 peptide substrate. Casp6N73T-transfected HEK293T cells showed elevated Casp6 mRNA levels similar to Casp6WT-transfected cells, but, in contrast to Casp6WT, did not accumulate active Casp6 subunits nor show increased Casp6 enzymatic activity. Electrophysiological and morphological assessments showed that Casp6N73T recombinant protein caused less neurofunctional damage and neurodegeneration in hippocampal CA1 pyramidal neurons than Casp6WT. Lastly, CASP6 mRNA levels were increased in several AD brain regions confirming the implication of Casp6 in AD. These studies suggest that the rare Casp6N73T variant may protect against hippocampal atrophy due to its altered catalysis of natural protein substrates and intracellular instability thus leading to less Casp6-mediated damage to neuronal structure and function.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , CA1 Region, Hippocampal/pathology , Caspase 6/genetics , Caspase 6/metabolism , Polymorphism, Single Nucleotide , Synaptic Transmission , Alzheimer Disease/enzymology , Amino Acid Substitution , Brain/enzymology , Brain/pathology , Caspase 1/genetics , Caspase 1/metabolism , Caspase 6/chemistry , Enzyme Precursors/metabolism , HEK293 Cells , Hippocampus , Humans , Lamin Type A/metabolism , Mutation, Missense , Nerve Degeneration , Pyramidal Cells/cytology , Pyramidal Cells/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Tubulin/metabolism
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.
Mol Immunol ; 132: 8-20, 2021 04.
Article in English | MEDLINE | ID: mdl-33524772

ABSTRACT

The cysteine-containing aspartate specific proteinase (caspase) family plays important roles in apoptosis and the maintenance of homeostasis in lampreys. We conducted genomic and functional comparisons of six distinct lamprey caspase groups with human counterparts to determine how these expanded molecules evolved to adapt to the changing caspase-mediated signaling pathways. Our results showed that lineage-specific duplication and rearrangement were responsible for expanding lamprey caspases 3 and 7, whereas caspases 1, 6, 8, and 9 maintained a relatively stable genome and protein structure. Lamprey caspase family molecules displayed various expression patterns and were involved in the innate immune response. Caspase 1 and 7 functioned as a pattern recognition receptor with a broad-spectrum of microbial recognition and bactericidal effect. Additionally, caspases 1 and 7 may induce cell apoptosis in a time- and dose-dependent manner; however, apoptosis was inhibited by caspase inhibitors. Thus, these molecules may reflect the original state of the vertebrates caspase family. Our phylogenetic and functional data provide insights into the evolutionary history of caspases and illustrate their functional characteristics in primitive vertebrates.


Subject(s)
Apoptosis/genetics , Caspases/genetics , Immunity, Innate , Lampreys/genetics , Signal Transduction/immunology , Animals , Apoptosis/drug effects , Caspase 1/chemistry , Caspase 1/genetics , Caspase 1/isolation & purification , Caspase 1/metabolism , Caspase 3/chemistry , Caspase 3/genetics , Caspase 3/metabolism , Caspase 6/chemistry , Caspase 6/genetics , Caspase 6/metabolism , Caspase 7/chemistry , Caspase 7/genetics , Caspase 7/isolation & purification , Caspase 7/metabolism , Caspase 8/chemistry , Caspase 8/genetics , Caspase 8/metabolism , Caspase 9/chemistry , Caspase 9/genetics , Caspase 9/metabolism , Caspase Inhibitors/pharmacology , Caspases/chemistry , Caspases/isolation & purification , Caspases/metabolism , Evolution, Molecular , Gene Duplication , Gene Rearrangement , Genome , Genomics , HeLa Cells , Humans , Immunity, Innate/genetics , Lampreys/growth & development , Lampreys/immunology , Lampreys/metabolism , Phylogeny , Recombinant Proteins , Sequence Alignment , Signal Transduction/genetics , Staphylococcus aureus/drug effects , Up-Regulation , Vibrio/drug effects
7.
J Enzyme Inhib Med Chem ; 36(1): 336-344, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33390035

ABSTRACT

The main purpose of this investigation was to evaluate the effect of anticancer active compounds (I-VIII) on zebrafish development in order to select the safest molecules. Larval mortality, embryo hatchability and malformations were end-points used to assess the acute toxicity among embryos and larvae from compounds-/pemetrexed-treated and control groups. LC50 and MNLC (maximal non-lethal concentration) were determined. Lipophilicity-dependent structure-toxicity relationships were established. The results clearly indicated that the majority of test molecules are safe for zebrafish individuals and simultaneously are less toxic than an anticancer agent - pemetrexed. The subsequent aim of this study was to elucidate the molecular mechanism of antiproliferative activity of the most selective compounds. Substantially increased activation of caspase-6 and -8 in cancerous cell lines confirmed the proapoptotic action of molecules examined. Considering the safety for zebrafish individuals, the title compounds as inducers of apoptosis are promising drug candidates in the preclinical phase of drug development.


Subject(s)
Antineoplastic Agents/pharmacology , Embryo, Nonmammalian/drug effects , Fluorocarbons/pharmacology , Larva/drug effects , Triazines/pharmacology , A549 Cells , Animals , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Caspase 6/genetics , Caspase 6/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor , Embryo, Nonmammalian/abnormalities , Embryo, Nonmammalian/metabolism , Fluorocarbons/chemical synthesis , Gene Expression/drug effects , HeLa Cells , Humans , Hydrophobic and Hydrophilic Interactions , Larva/anatomy & histology , Larva/growth & development , Larva/metabolism , Pemetrexed/toxicity , Structure-Activity Relationship , Toxicity Tests , Triazines/chemical synthesis , Zebrafish/growth & development
8.
Dev Comp Immunol ; 118: 103987, 2021 05.
Article in English | MEDLINE | ID: mdl-33359598

ABSTRACT

Caspases are highly conserved cysteine-dependent aspartyl-specific proteases that play an important role in regulating cell death and inflammation. However, the caspase genes have not been systematically studied in rainbow trout (Oncorhynchus mykiss). Rainbow trout experienced 4 rounds (4R) of genome duplication in the evolutionary history. Thereby an increased numbers of paralogs are observed in trout, probably with more complicated gene functions. We identified 18 caspase genes in rainbow trout, including two inflammatory caspases (casp1a, casp1b), six apoptosis executioner caspases (casp3, casp3a1, casp3a2, casp3b, casp6, and casp7), nine apoptosis initiator caspases (casp2a, casp2b, casp8, casp9a, casp9b, casp10a, casp10b, casp20a, and casp20b) and one uncategorized caspase gene (casp17). To investigate the potentially physiological functions of caspase genes, we challenged the rainbow trout with Aeromonas salmonicida (A. salmonicida) and Vibrio anguillarum (V. anguillarum). Results showed that the CASP3-regulated intrinsic apoptosis was activated after A. salmonicida infection, while the CASP8 and CASP6-regulated extrinsic apoptosis exerted the greatest effect on trout challenged with V. anguillarum. In response to V. anguillarum infection, the data of RNA-Seq further showed the casp8 was tightly integrated with the significantly enriched Gene Ontology terms and functional pathways, including apoptosis regulation, pathogen detection and immunomodulation. Our study provides a foundation for the physiological functions and regulatory network of the caspase genes in teleosts.


Subject(s)
Apoptosis/immunology , Caspase 8/genetics , Fish Diseases/immunology , Fish Proteins/genetics , Oncorhynchus mykiss/immunology , Aeromonas salmonicida/immunology , Animals , Apoptosis/genetics , Caspase 3/genetics , Caspase 3/metabolism , Caspase 6/genetics , Caspase 6/metabolism , Caspase 8/metabolism , Fish Diseases/microbiology , Fish Proteins/metabolism , Gene Duplication , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Oncorhynchus mykiss/genetics , Oncorhynchus mykiss/microbiology , Phylogeny , Synteny , Vibrio/immunology , Whole Genome Sequencing
9.
Cell Biochem Biophys ; 78(3): 291-299, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32592127

ABSTRACT

The predominance of Alzheimer's disease (AD) among the aged remains a global challenge. As such, the search for alternative and effective therapeutic options continuous unabated. Among the therapeutic targets explored over the years toward impeding the progression of AD is caspase-6 (Casp6), although selectively targeting Casp6 remains a challenge due to high homology with other members of the caspase family. Methyl 3-[(2,3-dihydro-1-benzofuran-2-yl formamido) methyl]-5-(furan-2-amido) benzoate (C13), a novel allosteric inhibitor, is reportedly shown to exhibit selective inhibition against mutant human Casp6 variants (E35K). However, structural and atomistic insights accounting for the reported inhibitory prowess of C13 remains unresolved. In this study, we seek to unravel the mechanistic selectivity of C13 coupled with the complementary effects of E35K single-nucleotide polymorphism (SNP) relative to Casp6 inhibition. Analyses of binding dynamics revealed that the variant Lysine-35 mediated consistent high-affinity interactions with C13 at the allosteric site, possibly forming the molecular basis of the selectivity of C13 as well as its high binding free energy as estimated. Analysis of residue interaction network around Glu35 and Lys35 revealed prominent residue network distortions in the mutant Casp6 conformation evidenced by a decrease in node degree, reduced number of edges and an increase short in path length relative to a more compact conformation in the wild system. The relatively higher binding free energy of C13 coupled with the stronger intermolecular interactions elicited in the mutant conformation further suggests that the mutation E35K probably favours the inhibitory activity of C13. Further analysis of atomistic changes showed increased C-α atom deviations consistent with structural disorientations in the mutant Casp6. Structural Insights provided could open up a novel paradigm of structure-based design of selective allosteric inhibition of Casp6 towards the treatment of neurodegenerative diseases.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Caspase 6/genetics , Caspase Inhibitors/pharmacology , Mutation , Polymorphism, Single Nucleotide , Allosteric Site , Caspase 6/chemistry , Drug Design , Humans , Imaging, Three-Dimensional , Molecular Dynamics Simulation , Protein Binding , Protein Conformation
10.
Vet Microbiol ; 242: 108610, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32122614

ABSTRACT

Reticuloendotheliosis virus (REV) infection of multiple avian species can lead to a number of diseases such as runting syndrome, immunosuppression and oncogenesis, causing major economic losses. MicroRNAs play important roles in post-transcriptional regulation, effectively inhibiting protein synthesis, and participating in many biological processes in cells, including proliferation, differentiation, apoptosis, lipometabolism, virus infection and replication, and tumorigenesis. Based on our previous high-throughput sequencing results, we explore the regulatory mechanisms of microRNA-155(miR-155) in chicken embryo fibroblasts (CEFs) in response to REV infection. Our results revealed expression of miR-155 in CEFs after REV infection upregulated in a time- and dose-dependent manner, indicating miR-155 plays a role in REV infection in CEFs indeed. After transfected with miR-155-mimic and miR-155-inhibitor, we found overexpression of miR-155 targeted caspase-6 and FOXO3a to inhibit apoptosis and accelerate cell cycle, thus improving viability of REV-infected CEFs. This result also verified the protective role of miR-155 in the viability of CEFs in the presence of REV. Knockdown of miR-155 also supported these above conclusions. Our findings uncover a new mechanism of REV pathogenesis in CEFs, and also provide a theoretical basis for uncovering new effective treatment and prevention methods for RE based on miR-155.


Subject(s)
Fibroblasts/virology , Gene Expression Regulation , Host Microbial Interactions/genetics , MicroRNAs/genetics , Reticuloendotheliosis virus/physiology , Retroviridae Infections/veterinary , Animals , Apoptosis , Caspase 6/genetics , Chick Embryo/virology , Gene Knockdown Techniques , High-Throughput Nucleotide Sequencing , Retroviridae Infections/genetics , Up-Regulation
11.
Int J Mol Sci ; 21(3)2020 Feb 09.
Article in English | MEDLINE | ID: mdl-32050445

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder and is the most common form of dementia in the elderly. Caspases, a family of cysteine proteases, are major mediators of apoptosis and inflammation. Caspase-6 is considered to be an up-stream modulator of AD pathogenesis as active caspase-6 is abundant in neuropil threads, neuritic plaques, and neurofibrillary tangles of AD brains. In order to further elucidate the role of caspase-6 activity in the pathogenesis of AD, we produced a double transgenic mouse model, combining the 5xFAD mouse model of AD with caspase-6 knock out (C6-KO) mice. Behavioral examinations of 5xFAD/C6-KO double transgenic mice showed improved performance in spatial learning, memory, and anxiety/risk assessment behavior, as compared to 5xFAD mice. Hippocampal mRNA expression analyses showed significantly reduced levels of inflammatory mediator TNF-α, while the anti-inflammatory cytokine IL-10 was increased in 5xFAD/C6-KO mice. A significant reduction in amyloid-ß plaques could be observed and immunohistochemistry analyses showed reduced levels of activated microglia and astrocytes in 5xFAD/C6-KO, compared to 5xFAD mice. Together, these results indicate a substantial role for caspase-6 in the pathology of the 5xFAD model of AD and suggest further validation of caspase-6 as a potential therapeutic target for AD.


Subject(s)
Alzheimer Disease/genetics , Caspase 6/genetics , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Animals , Disease Models, Animal , Female , Gene Deletion , Male , Memory , Mice , Mice, Knockout , Mutation , Plaque, Amyloid/genetics , Plaque, Amyloid/pathology , Plaque, Amyloid/physiopathology , Spatial Learning
12.
Gene Ther ; 26(6): 230-239, 2019 06.
Article in English | MEDLINE | ID: mdl-30962534

ABSTRACT

In order to adapt ex vivo regional gene therapy for clinical applications in orthopaedic surgery, safety issues must be considered. In this study we developed a suicide approach using a dual gene expression two step transcriptional amplification lentiviral vector (LV-TSTA) encoding BMP-2 and an inducible caspase 9 (iC9) system that selectively induces apoptosis upon activation with a chemical inducer of dimerization (CID). Transduction of rat bone marrow stromal cells (RBMSCs) with LV-TSTA-iC9/BMP-2 led to abundant BMP-2 production (90.3 ± 7.9 ng/24 h/106 cells) in vitro and stimulated bone formation in a mouse muscle pouch in the absence of CID. Moreover it was shown that CID could be used to selectively induce apoptosis in iC9-transduced cells both in vitro and in vivo. Double exposure to serial dilutions of CID decreased in vitro production of BMP-2 by 85-87% and Luc activity by 97-99% in iC9/BMP-2 or iC9/Luc-transduced cells respectively. Early administration of CID (Days 0-1 post-op) in mice implanted with iC9/BMP-2-transduced RBMSCs was effective in blocking bone formation, indicating that CID was toxic to the transduced cells. In iC9/Luc-implanted mice, late administration of two doses of CID (Days 27-28 post-op) significantly reduced the luciferase signal. The current study provides proof of concept for the potential clinical application of regulated gene therapy to promote bone repair.


Subject(s)
Apoptosis , Bone Regeneration , Caspase 6/genetics , Genetic Therapy/methods , Animals , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Caspase 6/metabolism , Cells, Cultured , Female , Genetic Vectors/genetics , Lentivirus/genetics , Mice , Mice, Inbred NOD , Protein Multimerization , Rats
13.
Sci Rep ; 9(1): 5504, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30940883

ABSTRACT

Caspase-6 is a cysteine protease that plays essential roles in programmed cell death, axonal degeneration, and development. The excess neuronal activity of Caspase-6 is associated with Alzheimer disease neuropathology and age-dependent cognitive impairment. Caspase-6 inhibition is a promising strategy to stop early stage neurodegenerative events, yet finding potent and selective Caspase-6 inhibitors has been a challenging task due to the overlapping structural and functional similarities between caspase family members. Here, we investigated how four rare non-synonymous missense single-nucleotide polymorphisms (SNPs), resulting in amino acid substitutions outside human Caspase-6 active site, affect enzyme structure and catalytic efficiency. Three investigated SNPs were found to align with a putative allosteric pocket with low sequence conservation among human caspases. Virtual screening of 57,700 compounds against the putative Caspase-6 allosteric pocket, followed by in vitro testing of the best virtual hits in recombinant human Caspase-6 activity assays identified novel allosteric Caspase-6 inhibitors with IC50 and Ki values ranging from ~2 to 13 µM. This report may pave the way towards the development and optimisation of novel small molecule allosteric Caspase-6 inhibitors and illustrates that functional characterisation of rare natural variants holds promise for the identification of allosteric sites on other therapeutic targets in drug discovery.


Subject(s)
Caspase 6/chemistry , Caspase 6/metabolism , Caspase Inhibitors/pharmacology , Mutation, Missense , Small Molecule Libraries/pharmacology , Allosteric Regulation/drug effects , Amino Acid Substitution , Caspase 6/genetics , Caspase Inhibitors/chemistry , Catalytic Domain , Computer Simulation , Crystallography, X-Ray , Humans , Models, Molecular , Polymorphism, Single Nucleotide , Protein Binding , Protein Conformation , Small Molecule Libraries/chemistry , Structure-Activity Relationship
14.
Free Radic Biol Med ; 134: 567-580, 2019 04.
Article in English | MEDLINE | ID: mdl-30769159

ABSTRACT

Recent reports in pathophysiology of neurodegenerative diseases (ND) have linked nuclear lamina degradation/deficits to neuronal cell death. Lamin-B1 damage is specifically involved in this process leading to nuclear envelope invagination and heterochromatin rearrangement. The underlying mechanisms involved in these events are not yet defined. In this study, while examining the effect of Thioredoxin-1(Trx1) inhibition on cell death in a model of oxidative stress, we noted robust nuclear invagination in SH-SY5Y cells. Evaluation of nuclear lamina proteins revealed lamin-B1 cleavage that was prevented by caspase-6 (CASP6) inhibitor and exacerbated after pharmacologic/genetic inhibition of Trx1 system, but not after glutathione depletion. Activation of CASP6 was upstream of CASP3/7 activation and its inhibition was sufficient to prevent cell death in our system. The effect of Trx1 redox status on CASP6 activation was assessed by administration of reduced/oxidized forms in cell-free nuclei preparation and purified enzymatic assays. Although reduced Trx1 decreased CASP6 enzymatic activity and lamin-B1 cleavage, the fully oxidized Trx1 showed opposite effects. The enhanced CASP6 activation was also associated with lower levels of DJ-1, a neuroprotective and master regulator of cellular antioxidants. The implication of our findings in ND pathophysiology was strengthened with detection of lower Trx1 levels in the hippocampi tissue of a mouse model of Alzheimer's disease. This coincided with higher CASP6 activation resulting in increased lamin-B1 and DJ-1 depletion. This study provides a first mechanistic explanation for the key regulatory role of Trx1 as a gatekeeper in activation of CASP6 and induction of nuclear invagination, an important player in ND pathophysiology.


Subject(s)
Alzheimer Disease/pathology , Antioxidants/metabolism , Caspase 6/metabolism , Neuroblastoma/pathology , Nuclear Lamina/pathology , Oxidative Stress , Thioredoxins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Caspase 6/genetics , Female , Glutathione , Humans , Male , Mice , Mice, Transgenic , Neuroblastoma/genetics , Neuroblastoma/metabolism , Nuclear Lamina/metabolism , Oxidation-Reduction , Thioredoxins/genetics , Tumor Cells, Cultured
15.
BMC Vet Res ; 15(1): 20, 2019 Jan 08.
Article in English | MEDLINE | ID: mdl-30621683

ABSTRACT

BACKGROUND: Caspases are a family of conserved intracellular cysteine-dependent aspartate-specific cysteine proteases that play important roles in regulating cell death and inflammation. Our previous study revealed the importance of the inflammatory caspase 1 gene in extracellular ATP-mediated immune signaling in Japanese flounder, Paralichthys olivaceus. To explore the potential roles of other caspases in P. olivaceus innate immunity, we extended our study by characterizing of the responses of four additional P. olivaceus caspase genes, termed JfCaspase 2, 3, 6 and 8, to inflammatory challenge and extracellular ATP stimulation. RESULTS: Sequence analysis revealed that the domain structures of all the Japanese flounder caspase proteins are evolutionarily conserved. Quantitative real-time PCR analysis showed that the JfCaspase 2, 3, 6 and 8 genes were expressed ubiquitously but at unequal levels in all examined Japanese flounder normal tissues. In addition, the basal gene expression levels of JfCaspase 2, 3, 6 and 8 were higher than those of JfCaspase 1 in both Japanese flounder head kidney macrophages (HKMs) and peripheral blood leukocytes (PBLs). Furthermore, immune challenge experiments showed that the inflammatory stimuli LPS and poly(I:C) significantly modulated the expression of the JfCaspase 2, 3, 6 and 8 genes in Japanese flounder immune cells. Finally, DNA fragmentation, associated with increased extracellular ATP-induced JfCaspase 2, 3, 6 and 8 gene expression and enzymatic activity, was inhibited by the caspase inhibitor Z-VAD-FMK in the HKMs. CONCLUSION: Our findings demonstrate broad participation of multiple caspase genes in response to inflammatory stimulation in Japanese flounder immune cells and provide new evidence for the involvement of caspase(s) in extracellular ATP-induced apoptosis in fish.


Subject(s)
Adenosine Triphosphate/pharmacology , Caspase 2/genetics , Caspase 3/genetics , Caspase 6/genetics , Caspase 8/genetics , Fish Proteins/genetics , Flounder/immunology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Caspase 2/physiology , Caspase 3/physiology , Caspase 6/physiology , Caspase 8/physiology , Fish Proteins/physiology , Flounder/genetics , Gene Expression Regulation/drug effects , Genes/drug effects , Immunity, Innate/drug effects , Immunity, Innate/immunology , Lipopolysaccharides/pharmacology , Phylogeny , Real-Time Polymerase Chain Reaction/veterinary , Sequence Alignment/veterinary , Sequence Analysis, DNA/veterinary
16.
J Biol Chem ; 294(1): 71-88, 2019 01 04.
Article in English | MEDLINE | ID: mdl-30420425

ABSTRACT

Caspases are cysteine-aspartic proteases involved in the regulation of programmed cell death (apoptosis) and a number of other biological processes. Despite overall similarities in structure and active-site composition, caspases show striking selectivity for particular protein substrates. Exosites are emerging as one of the mechanisms by which caspases can recruit, engage, and orient these substrates for proper hydrolysis. Following computational analyses and database searches for candidate exosites, we utilized site-directed mutagenesis to identify a new exosite in caspase-6 at the hinge between the disordered N-terminal domain (NTD), residues 23-45, and core of the caspase-6 structure. We observed that substitutions of the tri-arginine patch Arg-42-Arg-44 or the R44K cancer-associated mutation in caspase-6 markedly alter its rates of protein substrate hydrolysis. Notably, turnover of protein substrates but not of short peptide substrates was affected by these exosite alterations, underscoring the importance of this region for protein substrate recruitment. Hydrogen-deuterium exchange MS-mediated interrogation of the intrinsic dynamics of these enzymes suggested the presence of a substrate-binding platform encompassed by the NTD and the 240's region (containing residues 236-246), which serves as a general exosite for caspase-6-specific substrate recruitment. In summary, we have identified an exosite on caspase-6 that is critical for protein substrate recognition and turnover and therefore highly relevant for diseases such as cancer in which caspase-6-mediated apoptosis is often disrupted, and in neurodegeneration in which caspase-6 plays a central role.


Subject(s)
Caspase 6/chemistry , Mutation, Missense , Neoplasm Proteins/chemistry , Neoplasms/enzymology , Neurodegenerative Diseases/enzymology , Amino Acid Substitution , Arginine/chemistry , Arginine/genetics , Arginine/metabolism , Caspase 6/genetics , Caspase 6/metabolism , Humans , Hydrolysis , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Protein Domains
17.
Fish Shellfish Immunol ; 83: 406-409, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30240802

ABSTRACT

Apoptosis is a basic biological phenomenon of cells, which is an important component in the evolution of organisms, the stabilization of the internal environment and the development of multiple systems. In addition, the caspase protein family plays an important role in these pathways of apoptosis. Among them, apoptotic executors can directly act on specific substrates to complete the apoptotic response. In this study, we identified the Caspase-6 and Caspase-7 genes of miiuy croaker, and then analyzed the evolution of the whole Caspase family, furthermore described the evolutionary selection sites of the caspase-6 and caspase-7 genes in fish. The results showed that Caspase-6 gene appeared earlier than Caspase-7 in species evolution and gene duplication in teleost fish. Moreover, we also found that caspase-6 gene had no potential positive selection sites in the evolution of fish. Unlike the caspase-6 gene, the caspase-7 gene did not appear to be missed or replicated during the evolution of the species, while, it to be found two potential positive selection sites.


Subject(s)
Caspase 6/genetics , Caspase 7/genetics , Fish Proteins/genetics , Perciformes/genetics , Animals , Evolution, Molecular
18.
Fish Shellfish Immunol ; 80: 232-240, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29890217

ABSTRACT

In this study, a novel caspase-6 named HLcaspase-6 was identified from sea cucumber Holothuria leucospilota. The full-length cDNA of HLcaspase-6 is 2195 bp in size, containing a 126 bp 5'-untranslated region (UTR), a 1043 bp 3'-UTR and a 1026 bp open reading frame (ORF) encoding a protein of 341 amino acids with a deduced molecular weight of 38.57 kDa. HLcaspase-6 contains the common signatures of the caspase family, including the conserved pentapeptide motif QACRG, as well as the P20 and P10 domains. In addition, HLcaspase-6 contains a short pro-domain. HLcaspase-6 mRNA is ubiquitously expressed in all tissues examined, with the highest transcript level in the intestine, followed by coelomocytes. In in vitro experiments, the expression of HLcaspase-6 mRNA in coelomocytes was significantly up-regulated by lipopolysaccharides (LPS) or polyriboinosinic-polyribocytidylic acid [poly (I:C)] challenge, suggesting that HLcaspase-6 might play important roles in the innate immune defense of sea cucumber against bacterial and viral infections. Moreover, we further confirmed that overexpression of HLcaspase-6 could induce apoptosis and activate the p53 signal pathway.


Subject(s)
Caspase 6/genetics , Sea Cucumbers/genetics , Amino Acid Sequence , Animals , Apoptosis , Base Sequence , Caspase 6/immunology , Cloning, Molecular , DNA, Complementary/genetics , HEK293 Cells , Humans , Lipopolysaccharides/pharmacology , Phylogeny , Poly I-C/pharmacology , RNA, Messenger/metabolism , Sea Cucumbers/immunology , Sequence Alignment , Sequence Analysis, DNA , Tumor Suppressor Protein p53/metabolism
19.
Immunol Cell Biol ; 96(10): 1072-1082, 2018 11.
Article in English | MEDLINE | ID: mdl-29863787

ABSTRACT

The caspase (Casp) family of proteases regulate both lymphocyte apoptosis and activation. Here, we show that Casp6 regulates early B-cell development. One-week-old Casp6 knockout (Casp6 KO) mice have significantly more splenic B-cell subsets than wild-type (WT) mice. Adult Casp6 KO mice have normal levels of total splenic B cells but have increased numbers of B1a B cells and CD43+ "transitional" or splenic red pulp (RP) B cells. These results suggested that Casp6 may function to control B-cell numbers under nonhomeostatic conditions and during B-cell development. Consistent with this model, reconstitution of B cells was dysregulated in Casp6 KO mice after sublethal irradiation. Furthermore, bone marrow pro-B, pre-B and immature B-cell numbers were significantly higher in 1-week-old Casp6 KO mice than in 1-week-old WT mice. Casp6 KO pro-B cells proliferated more in response to IL-7 than WT pro-B cells, suggesting that Casp6 regulates early B-cell responses to IL-7. Indeed, adult and aged Casp6 KO mice had elevated numbers of IL-7αR+ Sca1+ precursors of common lymphoid progenitors, suggesting Casp6 may help regulate progenitors of B cells and early B-lineage cells. Casp6 regulates B-cell programs both during early development and after antigen stimulation in the periphery.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Caspase 6/metabolism , Immunomodulation , Animals , Animals, Newborn , Caspase 6/genetics , Interleukin-7/metabolism , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Lymphopoiesis , Male , Mice , Mice, Knockout , Peyer's Patches/immunology , Peyer's Patches/metabolism , Spleen/cytology , Spleen/immunology , Spleen/metabolism
20.
Sci Rep ; 8(1): 7231, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29739971

ABSTRACT

Broodiness in laying hens results in atrophy of the ovary and consequently decreases productivity. However, the regulatory mechanisms that drive ovary development remain elusive. Thus, we collected atrophic ovaries (AO) from 380-day-old broody chickens (BC) and normal ovaries (NO) from even-aged egg-laying hens (EH) for RNA sequencing. We identified 3,480 protein-coding transcripts that were differentially expressed (DE), including 1,719 that were down-regulated and 1,761 that were up-regulated in AO. There were 959 lncRNA transcripts that were DE, including 56 that were down-regulated and 903 that were up-regulated. Among the116 miRNAs that were DE, 79 were down-regulated and 37 were up-regulated in AO. Numerous DE protein-coding transcripts and target genes for miRNAs/lncRNAs were significantly enriched in reproductive processes, cell proliferation, and apoptosis pathways. A miRNA-intersection gene-pathway network was constructed by considering target relationships and correlation of the expression levels between ovary development-related genes and miRNAs. We also constructed a competing endogenous RNA (ceRNA) network by integrating competing relationships between protein-coding genes and lncRNA transcripts, and identified several lncRNA transcripts predicted to regulate the CASP6, CYP1B1, GADD45, MMP2, and SMAS2 genes. In conclusion, we discovered protein-coding genes, miRNAs, and lncRNA transcripts that are candidate regulators of ovary development in broody chickens.


Subject(s)
Atrophy/genetics , Avian Proteins/genetics , MicroRNAs/genetics , Ovary/metabolism , RNA, Long Noncoding/genetics , Animals , Apoptosis/genetics , Atrophy/metabolism , Atrophy/pathology , Avian Proteins/metabolism , Caspase 6/genetics , Caspase 6/metabolism , Cell Proliferation , Chickens , Consummatory Behavior/physiology , Cytochrome P-450 CYP1B1/genetics , Cytochrome P-450 CYP1B1/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , MicroRNAs/metabolism , Ovary/pathology , RNA, Long Noncoding/metabolism , Zygote , GADD45 Proteins
SELECTION OF CITATIONS
SEARCH DETAIL