Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Biochem J ; 478(13): 2681-2696, 2021 07 16.
Article in English | MEDLINE | ID: mdl-34156061

ABSTRACT

Apoptosis is a regulated form of cell death essential to the removal of unwanted cells. At its core, a family of cysteine peptidases named caspases cleave key proteins allowing cell death to occur. To do so, each caspase catalytic pocket recognizes preferred amino acid sequences resulting in proteolysis, but some also use exosites to select and cleave important proteins efficaciously. Such exosites have been found in a few caspases, notably caspase-7 that has a lysine patch (K38KKK) that binds RNA, which acts as a bridge to RNA-binding proteins favoring proximity between the peptidase and its substrates resulting in swifter cleavage. Although caspase-7 interaction with RNA has been identified, in-depth characterization of this interaction is lacking. In this study, using in vitro cleavage assays, we determine that RNA concentration and length affect the cleavage of RNA-binding proteins. Additionally, using binding assays and RNA sequencing, we found that caspase-7 binds RNA molecules regardless of their type, sequence, or structure. Moreover, we demonstrate that the N-terminal peptide of caspase-7 reduces the affinity of the peptidase for RNA, which translates into slower cleavages of RNA-binding proteins. Finally, employing engineered heterodimers, we show that a caspase-7 dimer can use both exosites simultaneously to increase its affinity to RNA because a heterodimer with only one exosite has reduced affinity for RNA and cleavage efficacy. These findings shed light on a mechanism that furthers substrate recognition by caspases and provides potential insight into its regulation during apoptosis.


Subject(s)
Apoptosis , Caspase 7/metabolism , Lysine/metabolism , RNA/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Caspase 7/chemistry , Caspase 7/genetics , HCT116 Cells , HEK293 Cells , Humans , Immunoblotting , Lysine/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Protein Binding , Protein Multimerization , Proteolysis , RNA/genetics , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Recombinant Proteins/metabolism , Substrate Specificity
2.
Proc Natl Acad Sci U S A ; 116(43): 21521-21528, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31586028

ABSTRACT

To achieve swift cell demise during apoptosis, caspases cleave essential proteins for cell survival and removal. In addition to the binding of preferred amino acid sequences to its substrate-binding pocket, caspase-7 also uses exosites to select specific substrates. 4 lysine residues (K38KKK) located in the N-terminal domain of caspase-7 form such an exosite and promote the rapid proteolysis of the poly(ADP-ribose) polymerase 1 (PARP-1), but the mechanism of recognition remains mostly unknown. In this study, we show that the overall positive charge of the exosite is the critical feature of this evolutionarily conserved binding site. Additionally, interaction with the caspase-7 exosite involves both the Zn3 and BRCT domains of PARP-1 and is mediated by RNA. Indeed, PARP-1 proteolysis efficacy is sensitive to RNase A and promoted by added RNA. Moreover, using affinity chromatography and gel shift assays, we demonstrate that caspase-7, but not caspase-3 or a caspase-7 with a mutated exosite, binds nucleic acids. Finally, we show that caspase-7 prefers RNA-binding proteins (RNA-BPs) as substrates compared to caspase-3 and that RNA enhances proteolysis by caspase-7 of many of these RNA-BPs. Thus, we have uncovered an unusual way by which caspase-7 selects and cleaves specific substrates.


Subject(s)
Caspase 7/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , RNA-Binding Proteins/metabolism , RNA/metabolism , Amino Acid Motifs , Animals , Caspase 7/chemistry , Caspase 7/genetics , Humans , Mice , Poly (ADP-Ribose) Polymerase-1/chemistry , Poly (ADP-Ribose) Polymerase-1/genetics , Protein Binding , Protein Domains , Proteolysis , RNA/genetics , RNA-Binding Proteins/genetics , Substrate Specificity
3.
Anal Bioanal Chem ; 413(20): 5085-5093, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34169347

ABSTRACT

The protein heterogeneity at the single-cell level has been recognized to be vital for an understanding of various life processes during animal development. In addition, the knowledge of accurate quantity of relevant proteins at cellular level is essential for appropriate interpretation of diagnostic and therapeutic results. Some low-copy-number proteins are known to play a crucial role during cell proliferation, differentiation, and also in apoptosis. The fate decision is often based on the concentration of these proteins in the individual cells. This is likely to apply also for caspases, cysteine proteases traditionally associated with cell death via apoptosis but recently being discovered also as important factors in cell proliferation and differentiation. The hypothesis was tested in bone-related cells, where modulation of fate from apoptosis to proliferation/differentiation and vice versa is particularly challenging, e.g., towards anti-osteoporotic treatments and anti-cancer strategies. An ultrasensitive and highly selective method based on bioluminescence photon counting was used to quantify activated caspase-3/7 in order to demonstrate protein-level heterogeneity in individual cells within one population and to associate quantitative measurements with different cell fates (proliferation, differentiation, apoptosis). The results indicate a gradual increase of caspase-3/7 activation from the proliferative status to differentiation (more than three times) and towards apoptosis (more than six times). The findings clearly support one of the putative key mechanisms of non-apoptotic functions of pro-apoptotic caspases based on fine-tuning of their activation levels.


Subject(s)
Caspase 3/chemistry , Caspase 3/metabolism , Caspase 7/chemistry , Caspase 7/metabolism , Osteoblasts/cytology , Animals , Apoptosis , Caspase 3/genetics , Caspase 7/genetics , Cell Differentiation , Cell Line , Cell Proliferation , Enzyme Activation , Mice , Osteoblasts/physiology
4.
J Biol Chem ; 292(12): 4885-4897, 2017 03 24.
Article in English | MEDLINE | ID: mdl-28154009

ABSTRACT

Caspases are cysteine aspartate proteases that are major players in key cellular processes, including apoptosis and inflammation. Specifically, caspase-6 has also been implicated in playing a unique and critical role in neurodegeneration; however, structural similarities between caspase-6 and other caspase active sites have hampered precise targeting of caspase-6. All caspases can exist in a canonical conformation, in which the substrate binds atop a ß-strand platform in the 130's region. This caspase-6 region can also adopt a helical conformation that has not been seen in any other caspases. Understanding the dynamics and interconversion between the helical and strand conformations in caspase-6 is critical to fully assess its unique function and regulation. Here, hydrogen/deuterium exchange mass spectrometry indicated that caspase-6 is inherently and dramatically more conformationally dynamic than closely related caspase-7. In contrast to caspase-7, which rests constitutively in the strand conformation before and after substrate binding, the hydrogen/deuterium exchange data in the L2' and 130's regions suggested that before substrate binding, caspase-6 exists in a dynamic equilibrium between the helix and strand conformations. Caspase-6 transitions exclusively to the canonical strand conformation only upon substrate binding. Glu-135, which showed noticeably different calculated pK a values in the helix and strand conformations, appears to play a key role in the interconversion between the helix and strand conformations. Because caspase-6 has roles in several neurodegenerative diseases, exploiting the unique structural features and conformational changes identified here may provide new avenues for regulating specific caspase-6 functions for therapeutic purposes.


Subject(s)
Caspase 6/metabolism , Caspase 6/chemistry , Caspase 7/chemistry , Caspase 7/metabolism , Enzyme Stability , Humans , Molecular Dynamics Simulation , Protein Binding , Protein Conformation , Protein Conformation, alpha-Helical , Protons
5.
Proteins ; 86(11): 1202-1210, 2018 11.
Article in English | MEDLINE | ID: mdl-30194780

ABSTRACT

The regulation of apoptosis is a tightly coordinated process and caspases are its chief regulators. Of special importance are the executioner caspases, caspase-3/7, the activation of which irreversibly sets the cell on the path of death. Dysregulation of apoptosis, particularly an increased rate of cell death lies at the root of numerous human diseases. Although several peptide-based inhibitors targeting the homologous active site region of caspases have been developed, owing to their non-specific activity and poor pharmacological properties their use has largely been restricted. Thus, we sought to identify FDA-approved drugs that could be repurposed as novel allosteric inhibitors of caspase-3/7. In this study, we virtually screened a catalog of FDA-approved drugs targeting an allosteric pocket located at the dimerization interface of caspase-3/7. From among the top-scoring hits we short-listed 5 compounds for experimental validation. Our enzymatic assays using recombinant caspase-3 suggested that 4 out of the 5 drugs effectively inhibited caspase-3 enzymatic activity in vitro with IC50 values ranging ~10-55 µM. Structural analysis of the docking poses show the 4 compounds forming specific non-covalent interactions at the allosteric pocket suggesting that these molecules could disrupt the adjacently-located active site. In summary, we report the identification of 4 novel non-peptide allosteric inhibitors of caspase-3/7 from among FDA-approved drugs.


Subject(s)
Allosteric Regulation/drug effects , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Caspase 3/metabolism , Caspase 7/metabolism , Caspase Inhibitors/pharmacology , Drug Repositioning , Allosteric Site/drug effects , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Caspase 3/chemistry , Caspase 7/chemistry , Caspase Inhibitors/chemistry , Drug Approval , Drug Repositioning/methods , HEK293 Cells , Humans , Molecular Docking Simulation
6.
Angew Chem Int Ed Engl ; 56(46): 14443-14447, 2017 11 13.
Article in English | MEDLINE | ID: mdl-28940929

ABSTRACT

The caspase family of cysteine proteases are highly sought-after drug targets owing to their essential roles in apoptosis, proliferation, and inflammation pathways. High-throughput screening efforts to discover inhibitors have gained little traction. Fragment-based screening has emerged as a powerful approach for the discovery of innovative drug leads. This method has become a central facet of drug discovery campaigns in the pharmaceutical industry and academia. A fragment-based drug discovery campaign against human caspase-7 resulted in the discovery of a novel series of allosteric inhibitors. An X-ray crystal structure of caspase-7 bound to a fragment hit and a thorough kinetic characterization of a zymogenic form of the enzyme were used to investigate the allosteric mechanism of inhibition. This work further advances our understanding of the mechanisms of allosteric control of this class of pharmaceutically relevant enzymes, and provides a new path forward for drug discovery efforts.


Subject(s)
Caspase 7/metabolism , Drug Discovery/methods , Allosteric Regulation/drug effects , Apoptosis , Caspase 7/chemistry , Catalytic Domain , Crystallography, X-Ray , Humans , Isotopes/chemistry , Kinetics , Solvents/chemistry , Surface Plasmon Resonance
7.
Nutr Cancer ; 68(7): 1210-24, 2016 10.
Article in English | MEDLINE | ID: mdl-27618154

ABSTRACT

ABSTACT Artemisia nilagirica (Clarke) is a widely used medicinal herb in Indian traditional system of medicine. Therefore, the present study was designed to evaluate the effects of A. nilagirica extracts/fractions on inhibition of proliferation and apoptosis in a human monocytic leukemia (THP-1) cell line. The crude extracts (A. nilagirica ethyl acetate extract [ANE] and A. nilagirica methanolic extract [ANA]) showed cytotoxic activity toward THP-1 cells with the IC50 values of 38.21 ± 7.37 and 132.41 ± 7.19 µg/ml, respectively. However, the cytotoxic activity of active fractions (ANE-B and ANM-9) obtained after column chromatography was found to be much more pronounced than their parent extracts. The IC50 values of ANE-B and ANM-9 were found to be 27.04 ± 2.54 µg/ml and 12.70 ± 4.79 µg/ml, respectively, suggesting greater susceptibility of the malignant cells. Cell cycle analysis and terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick-end-labeling (TUNEL) assay revealed that inhibition of cell growth by A. nilagirica fractions on THP-1 cells was mediated by apoptosis. Active fractions of A. nilagirica increased the expression levels of caspase-3, -7, and poly-ADP-ribose polymerase (PARP), a critical member of the apoptotic pathway. These results suggested that active fractions of A. nilagirica may play a promising role in growth suppression by inducing apoptosis in human monocytic leukemic cells via mitochondria-dependent and death receptor-dependent apoptotic pathways.


Subject(s)
Anticarcinogenic Agents/isolation & purification , Antineoplastic Agents, Phytogenic/isolation & purification , Apoptosis/drug effects , Artemisia/chemistry , Leukemia, Monocytic, Acute/drug therapy , Macrophages, Peritoneal/drug effects , Animals , Anticarcinogenic Agents/adverse effects , Anticarcinogenic Agents/chemistry , Anticarcinogenic Agents/pharmacology , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Biological Assay , Caspase 3/chemistry , Caspase 3/genetics , Caspase 3/metabolism , Caspase 7/chemistry , Caspase 7/genetics , Caspase 7/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , India , Inhibitory Concentration 50 , Leukemia, Monocytic, Acute/metabolism , Leukemia, Monocytic, Acute/pathology , Macrophages, Peritoneal/cytology , Mice, Inbred BALB C , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Plant Extracts/adverse effects , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Poly(ADP-ribose) Polymerases/chemistry , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , THP-1 Cells
8.
Proc Natl Acad Sci U S A ; 110(21): 8477-82, 2013 May 21.
Article in English | MEDLINE | ID: mdl-23650375

ABSTRACT

Procaspase-3 (P3) and procaspase-7 (P7) are activated through proteolytic maturation to form caspase-3 (C3) and caspase-7 (C7), respectively, which serve overlapping but nonredundant roles as the executioners of apoptosis in humans. However, it is unclear if differences in P3 and P7 maturation mechanisms underlie their unique biological functions, as the structure of P3 remains unknown. Here, we report structures of P3 in a catalytically inactive conformation, structures of P3 and P7 bound to covalent peptide inhibitors that reveal the active conformation of the zymogens, and the structure of a partially matured C7:P7 heterodimer. Along with a biochemical analysis, we show that P3 is catalytically inactive and matures through a symmetric all-or-nothing process. In contrast, P7 contains latent catalytic activity and matures through an asymmetric and tiered mechanism, suggesting a lower threshold for activation. Finally, we use our structures to design a selection strategy for conformation specific antibody fragments that stimulate procaspase activity, showing that executioner procaspase conformational equilibrium can be rationally modulated. Our studies provide a structural framework that may help to explain the unique roles of these important proapoptotic enzymes, and suggest general strategies for the discovery of proenzyme activators.


Subject(s)
Caspase 3/chemistry , Caspase 7/chemistry , Enzyme Precursors/chemistry , Protein Multimerization/physiology , Apoptosis/physiology , Caspase 3/genetics , Caspase 3/metabolism , Caspase 7/genetics , Caspase 7/metabolism , Enzyme Activation/physiology , Enzyme Precursors/genetics , Enzyme Precursors/metabolism , Humans , Protein Structure, Quaternary , Protein Structure, Tertiary , Structure-Activity Relationship
9.
Biochem J ; 461(2): 279-90, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24779913

ABSTRACT

Caspases play important roles during apoptosis, inflammation and proliferation. The high homology among family members makes selective targeting of individual caspases difficult, which is necessary to precisely define the role of these enzymes. We have selected caspase-7-specific binders from a library of DARPins (designed ankyrin repeat proteins). The DARPins D7.18 and D7.43 bind specifically to procaspase 7 and active caspase 7, but not to other members of the family. Binding of the DARPins does not affect the active enzyme, but interferes with its activation by other caspases. The crystal structure of the caspase 7-D7.18 complex elucidates the high selectivity and the mode of inhibition. Combining these caspase-7-specific DARPins with the previously reported caspase-3-inhibitory DARPin D3.4S76R reduces the activity of caspase 3 and 7 in double-transfected HeLa cells during apoptosis. In addition, these cells showed less susceptibility to TRAIL (tumour-necrosis-factor-related apoptosis-inducing ligand)-induced apoptosis in living cell experiments. D7.18 and D7.43 are therefore novel tools for in vitro studies on procaspase 7 activation as well as for clarifying the role of its activation in different cellular processes. If applied in combination with D3.4S76R, they represent an excellent instrument to increase our understanding of these enzymes during various cellular processes.


Subject(s)
Caspase 3/metabolism , Caspase 7/metabolism , Caspase Inhibitors/pharmacology , Nuclear Proteins/pharmacology , Ankyrin Repeat , Apoptosis/drug effects , Caspase 3/chemistry , Caspase 7/chemistry , Caspase Inhibitors/chemistry , HeLa Cells , Humans , Models, Molecular , Molecular Imaging , Nuclear Proteins/chemistry , Peptide Library , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology
10.
Proc Natl Acad Sci U S A ; 109(15): 5669-74, 2012 Apr 10.
Article in English | MEDLINE | ID: mdl-22451931

ABSTRACT

During apoptosis, hundreds of proteins are cleaved by caspases, most of them by the executioner caspase-3. However, caspase-7, which shares the same substrate primary sequence preference as caspase-3, is better at cleaving poly(ADP ribose) polymerase 1 (PARP) and Hsp90 cochaperone p23, despite a lower intrinsic activity. Here, we identified key lysine residues (K(38)KKK) within the N-terminal domain of caspase-7 as critical elements for the efficient proteolysis of these two substrates. Caspase-7's N-terminal domain binds PARP and improves its cleavage by a chimeric caspase-3 by ∼30-fold. Cellular expression of caspase-7 lacking the critical lysine residues resulted in less-efficient PARP and p23 cleavage compared with cells expressing the wild-type peptidase. We further showed, using a series of caspase chimeras, the positioning of p23 on the enzyme providing us with a mechanistic insight into the binding of the exosite. In summary, we have uncovered a role for the N-terminal domain (NTD) and the N-terminal peptide of caspase-7 in promoting key substrate proteolysis.


Subject(s)
Caspase 7/chemistry , Caspase 7/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Proteolysis , Amino Acid Motifs , Amino Acid Sequence , Apoptosis , Caspase 3/metabolism , Cell Line , Humans , Intramolecular Oxidoreductases/metabolism , Models, Molecular , Molecular Sequence Data , Prostaglandin-E Synthases , Protein Structure, Tertiary , Structure-Activity Relationship , Substrate Specificity
11.
J Biol Chem ; 288(35): 25154-25164, 2013 Aug 30.
Article in English | MEDLINE | ID: mdl-23867459

ABSTRACT

Most of the therapeutic antibodies approved for clinical use are full-size IgG1 molecules. The interaction of the IgG1 Fc with the neonatal Fc receptor (FcRn) plays a critical role in maintaining their long half-life. We have hypothesized that isolated Fc domains could be engineered to functionally mimic full-size IgG1 (nanoantibodies) but with decreased (10-fold) size. Here, we report for the first time the successful generation of a soluble, monomeric CH3 domain (mCH3). In contrast to the wild-type dimeric CH3, the mCH3 exhibited pH-dependent binding to FcRn similar to that of Fc. The binding free energy of mCH3 to FcRn was higher than that of isolated CH2 but lower than that of Fc. Therefore, CH3 may contribute a larger portion of the free energy of binding to FcRn than CH2. A fusion protein of mCH3 with an engineered antibody domain (m36.4) also bound to FcRn in a pH-dependent fashion and exhibited significantly higher neutralizing activity against HIV-1 than m36.4-Fc fusion proteins. The m36.4-mCH3 fusion protein was monomeric, stable, soluble, and expressed at a high level in Escherichia coli. We also found that engineering an additional disulfide bond in mCH3 remarkably increased its thermal stability, whereas the FcRn binding was not affected. These data suggest that mCH3 could not only help in the exploration of the dual mechanisms of the CH3 contribution to Fc functions (dimerization and FcRn interactions) but could also be used for the development of candidate therapeutics with optimized half-life, enhanced tissue penetration, access to sterically restricted binding sites, and increased therapeutic efficacy.


Subject(s)
Caspase 7/chemistry , Immunoglobulin G/chemistry , Caspase 7/biosynthesis , Caspase 7/genetics , Escherichia coli , Gene Expression , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin G/genetics , Protein Binding , Protein Engineering/methods , Protein Stability , Protein Structure, Tertiary , Receptors, Fc/chemistry , Receptors, Fc/genetics , Receptors, Fc/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics
12.
Proteins ; 82(4): 546-55, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24085488

ABSTRACT

Proteases are prototypes of multispecific protein-protein interfaces. Proteases recognize and cleave protein and peptide substrates at a well-defined position in a substrate binding groove and a plethora of experimental techniques provide insights into their substrate recognition. We investigate the caspase family of cysteine proteases playing a key role in programmed cell death and inflammation, turning caspases into interesting drug targets. Specific ligand binding to one particular caspase is difficult to achieve, as substrate specificities of caspase isoforms are highly similar. In an effort to rationalize substrate specificity of two closely related caspases, we investigate the substrate promiscuity of the effector Caspases 3 and 7 by data mining (cleavage entropy) and by molecular dynamics simulations. We find a strong correlation between binding site rigidity and substrate readout for individual caspase subpockets explaining more stringent substrate readout of Caspase 7 via its narrower conformational space. Caspase 3 subpockets S3 and S4 show elevated local flexibility explaining the more unspecific substrate readout of that isoform in comparison to Caspase 7. We show by in silico exchange mutations in the S3 pocket of the proteases that a proline residue in Caspase 7 contributes to the narrowed conformational space of the binding site. These findings explain the substrate specificities of caspases via a mechanism of conformational selection and highlight the crucial importance of binding site local dynamics in substrate recognition of proteases. Proteins 2014; 82:546-555. © 2013 Wiley Periodicals, Inc.


Subject(s)
Caspase 3/chemistry , Caspase 3/metabolism , Caspase 7/chemistry , Caspase 7/metabolism , Catalytic Domain , Apoptosis/genetics , Binding Sites/genetics , Caspase 3/genetics , Caspase 7/genetics , Databases, Protein , Inflammation/genetics , Models, Molecular , Molecular Dynamics Simulation , Mutation , Protein Binding , Protein Conformation , Protein Isoforms/genetics , Substrate Specificity
13.
Reproduction ; 148(2): 221-35, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24850868

ABSTRACT

AKT, also referred to as protein kinase B (PKB or RAC), plays a critical role in controlling cell survival and apoptosis. To gain insights into the mechanisms regulating sperm survival after ejaculation, the role of AKT was investigated in stallion spermatozoa using a specific inhibitor and a phosphoflow approach. Stallion spermatozoa were washed and incubated in Biggers-Whitten-Whittingham medium, supplemented with 1% polyvinyl alcohol (PVA) in the presence of 0 (vehicle), 10, 20 or 30 µM SH5, an AKT inhibitor. SH5 treatment reduced the percentage of sperm displaying AKT phosphorylation, with inhibition reaching a maximum after 1 h of incubation. This decrease in phosphorylation was attributable to either dephosphorylation or suppression of the active phosphorylation pathway. Stallion spermatozoa spontaneously dephosphorylated during in vitro incubation, resulting in a lack of a difference in AKT phosphorylation between the SH5-treated sperm and the control after 4 h of incubation. AKT inhibition decreased the proportion of motile spermatozoa (total and progressive) and the sperm velocity. Similarly, AKT inhibition reduced membrane integrity, leading to increased membrane permeability and reduced the mitochondrial membrane potential concomitantly with activation of caspases 3 and 7. However, the percentage of spermatozoa exhibiting oxidative stress, the production of mitochondrial superoxide radicals, DNA oxidation and DNA fragmentation were not affected by AKT inhibition. It is concluded that AKT maintains the membrane integrity of ejaculated stallion spermatozoa, presumably by inhibiting caspases 3 and 7, which prevents the progression of spermatozoa to an incomplete form of apoptosis.


Subject(s)
Caspase 3/chemistry , Caspase 7/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Semen Preservation , Sperm Motility , Spermatozoa/cytology , Animals , Apoptosis , Blotting, Western , Caspase 3/metabolism , Caspase 7/metabolism , Cell Proliferation , Cells, Cultured , Flow Cytometry , Horses , Immunoenzyme Techniques , Male , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Semen Analysis , Spermatozoa/metabolism
14.
Bioorg Med Chem ; 22(24): 6735-45, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25468037

ABSTRACT

A small molecule library of N-acyl-substituted 1,2-benzisothiazol-3-one derivatives has been synthesized and evaluated as inhibitors of caspase-3 and -7, in which some of them showed nanomolar potency against caspase-3 and -7 in vitro. Meanwhile, in 10 lM concentration, both compounds 24 and 25 showed significant protection against apoptosis in camptothecin-induced Jurkat T cells system. The docking studies predicted the interactions and binding modes of the synthesized inhibitors in the caspase-3 active site.


Subject(s)
Caspase 3/metabolism , Caspase Inhibitors/chemical synthesis , Thiazoles/chemistry , Apoptosis/drug effects , Binding Sites , Camptothecin/toxicity , Caspase 3/chemistry , Caspase 7/chemistry , Caspase 7/metabolism , Caspase Inhibitors/chemistry , Caspase Inhibitors/pharmacology , Catalytic Domain , Humans , Jurkat Cells , Molecular Docking Simulation , Protein Binding , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/pharmacology
15.
Int J Mol Sci ; 15(2): 2722-37, 2014 Feb 17.
Article in English | MEDLINE | ID: mdl-24549175

ABSTRACT

We previously noted that kaempferol, a flavonol present in vegetables and fruits, reduced cell cycle progression of HT-29 cells. To examine whether kaempferol induces apoptosis of HT-29 cells and to explore the underlying molecular mechanisms, cells were treated with various concentrations (0-60 µmol/L) of kaempferol and analyzed by Hoechst staining, Annexin V staining, JC-1 labeling of the mitochondria, immunoprecipitation, in vitro kinase assays, Western blot analyses, and caspase-8 assays. Kaempferol increased chromatin condensation, DNA fragmentation and the number of early apoptotic cells in HT-29 cells in a dose-dependent manner. In addition, kaempferol increased the levels of cleaved caspase-9, caspase-3 and caspase-7 as well as those of cleaved poly (ADP-ribose) polymerase. Moreover, it increased mitochondrial membrane permeability and cytosolic cytochrome c concentrations. Further, kaempferol decreased the levels of Bcl-xL proteins, but increased those of Bik. It also induced a reduction in Akt activation and Akt activity and an increase in mitochondrial Bad. Additionally, kaempferol increased the levels of membrane-bound FAS ligand, decreased those of uncleaved caspase-8 and intact Bid and increased caspase-8 activity. These results indicate that kaempferol induces the apoptosis of HT-29 cells via events associated with the activation of cell surface death receptors and the mitochondrial pathway.


Subject(s)
Antineoplastic Agents/toxicity , Apoptosis/drug effects , Kaempferols/toxicity , Amino Acid Chloromethyl Ketones/pharmacology , Caspase 3/chemistry , Caspase 3/metabolism , Caspase 7/chemistry , Caspase 7/metabolism , Caspase 8/chemistry , Caspase 8/metabolism , Caspase 9/chemistry , Caspase 9/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cytochromes c/metabolism , DNA Fragmentation/drug effects , HT29 Cells , Humans , Mitochondria/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , bcl-X Protein/metabolism
16.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 8): 1514-21, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23897474

ABSTRACT

Caspase-7 is expressed as a proenzyme and is activated by initiator caspases upon the transmission of cell-death signals. Despite extensive structural and biochemical analyses, many questions regarding the mechanism of caspase-7 activation remain unanswered. Caspase-7 is auto-activated during overexpression in Escherichia coli, even in the absence of initiator caspases, indicating that procaspase-7 has intrinsic catalytic activity. When variants of procaspase-7 with altered L2 loops were prepared, a variant with six inserted amino acids showed meaningful catalytic activity which was inhibited by Ac-DEVD-CHO. The kinetic constants of the procaspase-7 variant were determined and its three-dimensional structure was solved with and without bound inhibitor. The homodimeric procaspase-7 bound to the inhibitor revealed an asymmetry. One monomer formed a complete active site bound to the inhibitor in collaboration with the L2 loop from the other monomer, whereas the other monomer had an incomplete active site despite the bound inhibitor. Consequently, the two L2 loops in homodimeric procaspase-7 served as inherent L2 and L2' loops forming one complete active site. These data represent the first three-dimensional structure of a procaspase-7 variant bound to a specific inhibitor, Ac-DEVD-CHO, and provide insight into the folding mechanism during caspase-7 activation and the basal activity level of procaspase-7.


Subject(s)
Caspase 7/chemistry , Caspase 7/metabolism , Caspase Inhibitors/chemistry , Oligopeptides/chemistry , Base Sequence , Caspase 7/genetics , Catalytic Domain , Crystallography, X-Ray , Enzyme Precursors/chemistry , Escherichia coli/genetics , Kinetics , Models, Molecular , Molecular Sequence Data , Mutation , Protein Conformation
17.
J Biol Chem ; 286(25): 22291-9, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21555521

ABSTRACT

p21-activated kinase (PAK) 2, a member of the PAK family of serine/threonine protein kinases, plays an important role in physiological processes such as motility, survival, mitosis, and apoptosis. However, the role of PAK2 in resistance to chemotherapy is unclear. Here we report that PAK2 is highly expressed in human breast cancer cell lines and human breast invasive carcinoma tissue compared with a human non-tumorigenic mammary epithelial cell line and adjacent normal breast tissue, respectively. Interestingly, we found that PAK2 can bind with caspase-7 and phosphorylate caspase-7 at the Ser-30, Thr-173, and Ser-239 sites. Functionally, the phosphorylation of caspase-7 decreases its activity, thereby inhibiting cellular apoptosis. Our data indicate that highly expressed PAK2 mediates chemotherapeutic resistance in human breast invasive ductal carcinoma by negatively regulating caspase-7 activity.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/pathology , Caspase 7/metabolism , Drug Resistance, Neoplasm , p21-Activated Kinases/metabolism , Amino Acid Sequence , Antineoplastic Agents/therapeutic use , Apoptosis/genetics , Base Sequence , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Carcinoma, Ductal/drug therapy , Carcinoma, Ductal/enzymology , Carcinoma, Ductal/metabolism , Carcinoma, Ductal/pathology , Caspase 7/chemistry , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Phosphorylation/drug effects , Protein Transport , p21-Activated Kinases/deficiency , p21-Activated Kinases/genetics
18.
J Comput Chem ; 33(24): 1927-35, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22648914

ABSTRACT

A homology model builder using simple restraining potentials based on spline-interpolated quadratic functions is developed and interfaced with CHARMM package. The continuity and stability of the potential function were validated, and the parameters were optimized using the CASP7 targets. The performance of the model builder was benchmarked to the Modeller program using the template-based modeling targets in CASP9. The benchmark results show that, while our builder yields the structures with slightly lower packing, backbone, and template modeling scores, our models show much better protein-like scores in terms of normalized discrete optimized protein energy, dipolar distance-scaled finite-ideal gas reference, Molprobity clash, Ramachandran appearance Z-score, and rotamer Z-score. As our model builder is interfaced with CHARMM, it is advantageous to directly use other CHARMM functionality and energy functions to refine the model structures or to use the models for other computational studies using CHARMM.


Subject(s)
Caspase 7/chemistry , Caspase 9/chemistry , Proteomics/methods , Sequence Homology, Amino Acid , Software , Algorithms , Amino Acid Sequence , Databases, Protein , Humans , Models, Molecular , Molecular Sequence Data , Thermodynamics
19.
Biol Chem ; 393(9): 933-41, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22944693

ABSTRACT

Many peptidases are thought to require non-active site interaction surfaces, or exosites, to recognize and cleave physiological substrates with high specificity and catalytic efficiency. However, the existence and function of protease exosites remain obscure owing to a lack of effective methods to identify and characterize exosite-interacting substrates. To address this need, we modified the cellular libraries of peptide substrates (CLiPS) methodology to enable the discovery of exosite-interacting peptide ligands. Invariant cleavage motifs recognized by the active sites of thrombin and caspase-7 were displayed on the outer surface of bacteria adjacent to a candidate exosite-interacting peptide. Exosite peptide libraries were then screened for ligands that accelerate cleavage of the active site recognition motif using two-color flow cytometry. Exosite CLiPS (eCLiPS) identified exosite-binding peptides for thrombin that were highly similar to a critical exosite interaction motif in the thrombin substrate, protease-activated receptor 1. Protease activity probes incorporating exosite-binding peptides were cleaved ten-fold faster than substrates without exosite ligands, increasing their sensitivity to thrombin activity in vitro. For comparison, screening with caspase-7 yielded peptides that modestly enhanced (two-fold) substrate cleavage rates. The eCLiPS method provides a new tool to profile the ligand specificity of protease exosites and to develop improved substrates.


Subject(s)
Peptides/chemistry , Peptides/metabolism , Thrombin/chemistry , Thrombin/metabolism , Amino Acid Sequence , Caspase 7/chemistry , Caspase 7/metabolism , Catalytic Domain , Humans , Kinetics , Ligands , Models, Molecular , Molecular Sequence Data , Receptor, PAR-1/chemistry , Receptor, PAR-1/metabolism , Substrate Specificity
20.
J Immunol ; 185(6): 3127-30, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20713892

ABSTRACT

Caspase-mediated cleavage of the DNA damage sensor poly(ADP-ribose) polymerase 1 (PARP1) is a hallmark of apoptosis. However, it remains unclear whether PARP1 is processed during pyroptosis, a specialized cell-death program that occurs upon activation of caspase-1 in inflammasome complexes. In this article, we show that activation of the Nlrp3 and Nlrc4 inflammasomes induces processing of full-length PARP1 into a fragment of 89 kDa in a stimulus-dependent manner. Macrophages deficient for caspase-1 and those lacking the inflammasome adaptors Nlrp3, Nlrc4, and ASC were highly resistant to cleavage, whereas macrophages lacking the downstream inflammasome effector caspase-7 were partially protected. A modest, but statistically significant, reduction in Nlrp3 inflammasome-induced pyroptosis was observed in PARP1 knockout macrophages. Thus, protease-mediated inactivation of PARP1 is a shared feature of apoptotic, necrotic, and pyroptotic cells.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Calcium-Binding Proteins/physiology , Carrier Proteins/physiology , Inflammation Mediators/physiology , Inflammation/immunology , Inflammation/pathology , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Animals , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Carrier Proteins/genetics , Caspase 1/chemistry , Caspase 1/physiology , Caspase 7/chemistry , Caspase 7/physiology , Cell Death/genetics , Cell Death/immunology , Cells, Cultured , Inflammation/enzymology , Inflammation Mediators/chemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/chemistry , Protein Processing, Post-Translational/immunology , Substrate Specificity/immunology
SELECTION OF CITATIONS
SEARCH DETAIL