Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
FEBS J ; 290(14): 3580-3594, 2023 07.
Article in English | MEDLINE | ID: mdl-36853170

ABSTRACT

The development of neurological pathologies is linked to the accumulation of protein aggregates like alpha-synuclein in Parkinson's disease and tau protein in Alzheimer's disease. Mono- or di-ubiquitination of these molecules has been reported to stabilize aggregates and contribute to the disorders. STIP1 Homologous and U-Box-containing protein 1 (STUB1) is a multifunctional protein that maintains proteostasis and insulin signalling. In spinocerebellar ataxia 16 (SCAR16), an autosomal recessive neurodegenerative disease, mutations in and aggregation of STUB1 are reported. Despite the well-accepted neuroprotective role of STUB1, very little is known of regulatory mechanisms that control the dynamics of STUB1 aggregate assembly. Here, we report that CARP2, a ubiquitin ligase, is a novel regulator of STUB1. CARP2 interacts and mono-ubiquitinates STUB1. Furthermore, we found that CARP2 regulates STUB1 through its TPR motif, a domain that is also associated with HSP70. Modification of STUB1 by CARP2 leads to detergent-insoluble aggregate formation. Importantly, pathogenic mutants of STUB1 are more prone than the wild-type to CARP2-mediated aggregate assembly. Hence our findings revealed CARPs (CARP1 & CARP2) as novel regulators of STUB1 and controlled its cytosolic versus aggregate dynamics.


Subject(s)
Carps , Neurodegenerative Diseases , Animals , Humans , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Kinetics , Carps/metabolism , Ubiquitination
2.
Autophagy ; 18(12): 2851-2864, 2022 12.
Article in English | MEDLINE | ID: mdl-35373701

ABSTRACT

Mutations in the ubiquitin ligase PRKN (parkin RBR E3 ubiquitin protein ligase) are associated with Parkinson disease and defective mitophagy. Conceptually, PRKN-dependent mitophagy is classified into two phases: 1. PRKN recruits to and ubiquitinates mitochondrial proteins; 2. formation of phagophore membrane, sequestering mitochondria for degradation. Recently, endosomal machineries are reported to contribute to the later stage for membrane assembly. We reported a role for endosomes in the events upstream of phase 1. We demonstrate that the endosomal ubiquitin ligase RFFL (ring finger and FYVE like domain containing E3 ubiquitin protein ligase) associated with damaged mitochondria, and this association preceded that of PRKN. RFFL interacted with PRKN, and stable recruitment of PRKN to damaged mitochondria was substantially reduced in RFFL KO cells. Our study unraveled a novel role of endosomes in modulating upstream pathways of PRKN-dependent mitophagy initiation.Abbreviations CCCP: carbonyl cyanide 3-chlorophenylhydrazone; DMSO: dimethyl sulfoxide; EGFP: enhanced green fluorescence protein; KO: knockout; PRKN: parkin RBR E3 ubiquitin protein ligase; RFFL: ring finger and FYVE like domain containing E3 ubiquitin protein ligase; UQCRC1: ubiquinol-cytochrome c reductase core protein 1; WT: wild-type.


Subject(s)
Autophagy , Protein Kinases , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Mitochondria/metabolism , Ubiquitin/metabolism , Endosomes/metabolism
3.
J Mol Biol ; 433(15): 167084, 2021 07 23.
Article in English | MEDLINE | ID: mdl-34081983

ABSTRACT

Long-term memory storage is modulated by the prion nature of CPEB3 forming the molecular basis for the maintenance of synaptic facilitation. Here we report that the first prion sub-domain PRD1 of mouse CPEB3 can autonomously form amyloid fibrils in vitro and punctate-like structures in vivo. A ninety-four amino acid sequence within the PRD1 domain, PRD1-core, displays high propensity towards aggregation and associated amyloid characteristics. PRD1-core is characterized using electron microscopy, X-ray diffraction, and solution-state NMR deuterium exchange experiments. Secondary structure elements deduced from solid-state NMR reveal a ß-rich core comprising of forty amino acids at the N-terminus of PRD1-core. The synthesized twenty-three amino acid long peptide containing the longest rigid segment (E124-H145) of the PRD1-core rapidly self-aggregates and forms fibrils, indicating a limited aggregation-prone region that could potentially activate the aggregation of the full-length protein. This study provides the first step in identifying the structural trigger for the CPEB3 aggregation process.


Subject(s)
Amyloid/metabolism , Memory, Long-Term , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Amino Acid Sequence , Animals , Cell Line , Circular Dichroism , Deuterium Exchange Measurement , Mice , Microscopy, Electron, Transmission , Protein Conformation, beta-Strand , Protein Domains , RNA-Binding Proteins/metabolism , X-Ray Diffraction
4.
Free Radic Biol Med ; 161: 198-211, 2020 12.
Article in English | MEDLINE | ID: mdl-33065180

ABSTRACT

The redox-active transition metals such as copper, iron, chromium, vanadium, and silica are known for its ROS generation via mechanisms such as Haber-Weiss and Fenton-type reactions. Nanoparticles of these metals induce oxidative stress due to acellular factors owing to their small size and more reactive surface area, leading to various cellular responses. The intrinsic enzyme-like activity of nano vanadium has fascinated the scientific community. However, information concerning their cellular uptake and time-dependent induced effects on their cellular organelles and biological activity is lacking. This comprehensive study focuses on understanding the precise molecular interactions of vanadium pentoxide nanoparticles (VnNp) and evaluate their specific "nano" induced effects on MDA-MB-231 cancer cells. Understanding the mechanism behind NP-induced ROS generation could help design a model for selective NP induced toxicity, useful for cancer management. The study demonstrated the intracellular persistence of VnNp and insights into its molecular interactions with various organelles and its overall effects at the cellular level. Where triple-negative breast cancer MDA-MB-231 cells resulted in 59.6% cell death towards 48 h of treatment and the normal fibroblast cells showed only 15.4% cell death, indicating an inherent anticancer property of VnNp. It acts as an initial reactive oxygen species quencher, by serving itself as an antioxidant, while; it was also found to alter the cellular antioxidant system with prolonged incubation. The VnNp accumulated explicitly in the lysosomes and mitochondria and modulated various cellular processes including impaired lysosomal function, mitochondrial damage, and autophagy. At more extended time points, VnNp influenced cell cycle arrest, inhibited cell migration, and potentiated the onset of apoptosis. Results are indicative of the fact that VnNp selectively induced breast cancer cell death and hence could be developed as a future drug molecule for breast cancer management. This could override the most crucial challenge of chemo-resistance that still remain as the main hurdle to cancer therapy.


Subject(s)
Autophagy , Nanoparticles , Apoptosis , Humans , Oxidation-Reduction , Reactive Oxygen Species , Vanadium Compounds
5.
J Cell Sci ; 133(11)2020 06 11.
Article in English | MEDLINE | ID: mdl-32409562

ABSTRACT

Lysosomal exocytosis and resealing of damaged plasma membrane are essential for cellular homeostasis and tumor invasion. However, very little is known of the molecular machinery that regulates these physiological processes. Moreover, no mutations in any of the known regulators of lysosomal exocytosis in primary tumors of patients have been characterized. Here we demonstrate that RNF167-a, a lysosomal-associated ubiquitin ligase, negatively regulates lysosomal exocytosis by inducing perinuclear clustering of lysosomes. Importantly, we also characterized a set of novel natural mutations in RNF167-a, which are commonly found in diverse tumor types. We found that RNF167-a-K97N mutant, unlike the wild type, localizes in the cytoplasm and does not promote perinuclear lysosomal clustering. Furthermore, cells expressing RNF167-a-K97N exhibit dispersed lysosomes, increased exocytosis and enhanced plasma membrane repair. Interestingly, these functional features of RNF167-a-K97N were shared with a naturally occurring short version of RNF167 (isoform RNF167-b). In brief, the results presented here reveal a novel role of RNF167-a, as well as its natural variants RNF167-a-K97N and RNF167-b, as an upstream regulator of lysosomal exocytosis and plasma membrane resealing.


Subject(s)
Exocytosis , Lysosomes , Cell Membrane , Humans
6.
Mol Cell ; 44(4): 511-2, 2011 Nov 18.
Article in English | MEDLINE | ID: mdl-22099299

ABSTRACT

In this issue of Molecular Cell, Skaug et al. (2011) propose a polyubiquitin-dependent, noncatalytic mechanism by which the deubiquitinase A20 inhibits IκB kinase and NF-κB activation.

7.
Autophagy ; 7(5): 552-4, 2011 May.
Article in English | MEDLINE | ID: mdl-21412052

ABSTRACT

Autophagy plays an evolutionarily conserved role in host defense against pathogens. Autophagic protection mechanisms against microbes range from regulating immune signaling responses to directly targeting the pathogens for lysosomal degradation. Toll-like receptors (TLRs) that detect conserved molecular features shared by pathogens regulate several innate immune responses including autophagy. Our recent study demonstrates that autophagy reported in response to TLR4-stimulation in macrophages is selective autophagy of aggresome-like induced structures (ALIS), and p62 (also known as SQSTM1) plays an essential role in this process. Treatment of macrophages with either Escherichia coli or lipopolysaccharide (LPS) results in the activation of nuclear factor erythroid 2-related factor 2 (Nrf2), leading to an increase in the levels of p62 mRNA and protein, assembly of ALIS and their autophagic degradation. This study revealed a signaling role for p62, distinct from its known function as a bacterial-targeting factor, which might be critical for cellular stress response during infection.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Autophagy/genetics , Inclusion Bodies/physiology , Macrophages/metabolism , Macrophages/physiology , Toll-Like Receptor 4/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autophagy/drug effects , Autophagy/physiology , Humans , Immunity, Innate/drug effects , Immunity, Innate/genetics , Immunity, Innate/physiology , Inclusion Bodies/drug effects , Inclusion Bodies/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Models, Biological , Sequestosome-1 Protein , Toll-Like Receptor 4/genetics
8.
Proc Natl Acad Sci U S A ; 108(4): 1427-32, 2011 Jan 25.
Article in English | MEDLINE | ID: mdl-21220332

ABSTRACT

Toll-like receptors (TLRs) play a crucial role in several innate immune responses by regulating autophagy, but little is known about how TLR signaling controls autophagy. Here we demonstrate that p62/SQSTM1 is required for TLR4-mediated autophagy, which we show as selective autophagy of aggresome-like induced structures (ALIS). Treatment with LPS or Escherichia coli induced LC3(+) dot-like structures, and their assembly, but not lysosomal degradation, occurred independently of classic autophagic machinery. Microscopic and ultrastructural analyses showed that p62 is a component of the induced LC3(+) dots and these TLR4-induced p62(+) structures resemble ALIS. The levels of p62 mRNA and protein were increased in TLR4-activated cells and knockdown of p62 suppressed the ALIS formation and LC3-II conversion. The accumulation of p62 and ALIS required activation of Nrf2 by reactive oxygen species-p38 axis-dependent TLR4/MyD88 signaling, suggesting a link between innate immune and oxidative-stress responses. These findings indicate that TLR4-driven induction of p62 plays an essential role in the formation and the autophagic degradation of ALIS, which might be critical for regulating host defense.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy , Heat-Shock Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Toll-Like Receptor 4/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Autophagy-Related Protein 7 , Cell Line , Cells, Cultured , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Heat-Shock Proteins/genetics , Humans , Immunoblotting , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/ultrastructure , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Immunoelectron , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NF-E2-Related Factor 2/genetics , RNA Interference , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequestosome-1 Protein , Toll-Like Receptor 4/genetics
9.
Apoptosis ; 16(2): 162-73, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21107705

ABSTRACT

Cells that form vascular system employ different mechanisms to offset deleterious consequences of exposure to cytokines and cells present in blood. Vascular homeostasis is sustained in part by genes, whose expression increases in response to hemodynamic forces in these cells. PP1201 (also known as RECS1) is one such gene whose expression level increases in response to laminar shear stress. Aged mice deficient in PP1201 are prone to develop cystic medial degeneration (CMD), a form of aortic aneurism manifested with loss of smooth muscle cells and accumulation of basophilic substances. Here we found that higher levels of PP1201 can protect against Fas ligand (FasL)-induced apoptosis. PP1201 interacted with the Fas receptor (CD95/Apo1) and colocalized with it in the Golgi compartment. Unlike its homolog lifeguard (LFG), PP1201 overexpression in several types of cells including primary human aortic smooth muscle cells (AoSMC) decreased the expression of Fas on the plasma membrane without changing the total Fas levels. Only high but not constitutive level of PP1201 controls Fas signaling. Our data suggest that PP1201 functions as an anti-apoptotic protein and its increased expression in vascular cells can contribute to homeostasis by reducing Fas trafficking to the cell membrane.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Cell Membrane/metabolism , Membrane Proteins/metabolism , fas Receptor/metabolism , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , Blood Vessels/cytology , Blotting, Western , Caspases/metabolism , Cell Line, Tumor , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Flow Cytometry , Golgi Apparatus/metabolism , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Stress, Physiological , fas Receptor/genetics
11.
Results Probl Cell Differ ; 49: 87-114, 2009.
Article in English | MEDLINE | ID: mdl-19582409

ABSTRACT

Death receptors are a subset of the tumor necrosis factor receptor (TNFR) family of proteins and share a characteristic cytoplasmic motif called the "death domain". In addition to mediating cell death, these receptors regulate cell proliferation, inflammatory responses, and tumor progression. Receptor occupancy triggers the assembly of several cytoplasmic molecules into distinct complexes, each initiating separate signaling events leading to different biological responses. Post-translational modifications involving ubiquitin, a peptide of 76 amino acids, regulate events at nearly all stages of signaling. All ubiquitin chains function as docking platforms for molecules with specific recognition motifs that either propagate the signal or target the protein for proteasomal degradation. Moreover, enzymes with ubiquitin thioesterase activity (deubiquitinating enzymes, or DUBs) reverse modifications by removing the ubiquitin chains, allowing ubiquitin editing at the molecular level. Ubiquitin protein ligases (E3s), DUBs, and signaling molecules with ubiquitin recognition motifs control TNFR1 mediated cell death and activation of NF-kappaB and JNK. Here, we discuss the current understanding of how these proteins regulate TNFR1 signaling.


Subject(s)
Protein Processing, Post-Translational/immunology , Receptors, Tumor Necrosis Factor, Type I/immunology , Signal Transduction/immunology , Ubiquitin/immunology , Ubiquitination/immunology , Amino Acid Motifs/immunology , Animals , Cell Death/immunology , Humans , Inflammation/immunology , MAP Kinase Kinase 4/immunology , NF-kappa B/immunology , Neoplasms/immunology , Ubiquitin Thiolesterase/immunology , Ubiquitin-Protein Ligases/immunology
12.
Curr Biol ; 18(9): 641-9, 2008 May 06.
Article in English | MEDLINE | ID: mdl-18450452

ABSTRACT

BACKGROUND: The proinflammatory cytokine tumor necrosis factor-alpha (TNF-alpha) elicits cellular responses by signaling through a receptor complex that includes the essential adaptor molecule RIP. One important consequence of signaling is activation of the transcription factor NF-kappaB, and failure to downregulate TNF-induced NF-kappaB transcriptional activity results in chronic inflammation and death. Internalization of the receptor complex plays an important regulatory role in TNF signaling. RESULTS: We report that CARP-2, a RING domain-containing ubiquitin protein ligase (E3), is a negative regulator of TNF-induced NF-kappaB activation. By virtue of its phospholipid-binding FYVE domain, CARP-2 localized to endocytic vesicles, where it interacted with internalized TNF-receptor complex, resulting in RIP ubiquitination and degradation. Knockdown of CARP-2 stabilized TNFR1-associated polyubiquitinated RIP levels after TNF simulation and enhanced activation of NF-kappaB. CONCLUSIONS: CARP-2 acts at the level of endocytic vesicles to limit the intensity of TNF-induced NF-kappaB activation by the regulated elimination of a necessary signaling component within the receptor complex.


Subject(s)
NF-kappa B/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Transport Vesicles/enzymology , Tumor Necrosis Factor-alpha/metabolism , Cell Line , Endocytosis/physiology , Humans , Ubiquitin-Protein Ligases/metabolism
13.
Mol Cell ; 30(2): 123-35, 2008 Apr 25.
Article in English | MEDLINE | ID: mdl-18439892

ABSTRACT

Originally described in insect viruses, cellular proteins with Baculoviral IAP repeat (BIR) motifs have been thought to function primarily as inhibitors of apoptosis. The subsequent finding that a subset of IAPs that contain a RING domain have ubiquitin protein ligase (E3) activity implied the presence of other functions. It is now known that IAPs are involved in mitotic chromosome segregation, cellular morphogenesis, copper homeostasis, and intracellular signaling. Here, we review the current understanding of the roles of IAPs in apoptotic and nonapoptotic processes and explore the notion that the latter represents the primary physiologic activities of IAPs.


Subject(s)
Apoptosis , Inhibitor of Apoptosis Proteins/chemistry , Inhibitor of Apoptosis Proteins/metabolism , Amino Acid Motifs , Animals , Cell Movement , Copper/metabolism , Humans , Immunity, Innate , Inhibitor of Apoptosis Proteins/genetics , Mice , Morphogenesis , Neoplasms/metabolism , Signal Transduction
14.
Nat Cell Biol ; 8(4): 398-406, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16547522

ABSTRACT

The transcription factor NF-kappaB is sequestered in the cytoplasm in a complex with IkappaB. Almost all NF-kappaB activation pathways converge on IkappaB kinase (IKK), which phosphorylates IkappaB resulting in Lys 48-linked polyubiquitination of IkappaB and its degradation. This allows migration of NF-kappaB to the nucleus where it regulates gene expression. IKK has two catalytic subunits, IKKalpha and IKKbeta, and a regulatory subunit, IKKgamma or NEMO. NEMO is essential for NF-kappaB activation, and NEMO dysfunction in humans is the cause of incontinentia pigmenti and hypohidrotic ectodermal dysplasia and immunodeficiency (HED-ID). The recruitment of IKK to occupied cytokine receptors, and its subsequent activation, are dependent on the attachment of Lys 63-linked polyubiquitin chains to signalling intermediates such as receptor-interacting protein (RIP). Here, we show that NEMO binds to Lys 63- but not Lys 48-linked polyubiquitin, and that single point mutations in NEMO that prevent binding to Lys 63-linked polyubiquitin also abrogates the binding of NEMO to RIP in tumour necrosis factor (TNF)-alpha-stimulated cells, the recruitment of IKK to TNF receptor (TNF-R) 1, and the activation of IKK and NF-kappaB. RIP is also destabilized in the absence of NEMO binding and undergoes proteasomal degradation in TNF-alpha-treated cells. These results provide a mechanism for NEMO's critical role in IKK activation, and a key to understanding the link between cytokine-receptor proximal signalling and IKK and NF-kappaB activation.


Subject(s)
Biosensing Techniques , I-kappa B Kinase/genetics , Lysine/metabolism , NF-kappa B/metabolism , Ubiquitin/metabolism , Electrophoretic Mobility Shift Assay , HeLa Cells , Humans , I-kappa B Kinase/metabolism , Immunoprecipitation , Lysine/genetics , NF-kappa B/genetics , Point Mutation , Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases , Receptors, Tumor Necrosis Factor, Type I/metabolism , Saccharomyces cerevisiae , Signal Transduction , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Two-Hybrid System Techniques
15.
PLoS Biol ; 3(6): e183, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15941357

ABSTRACT

Caspases are responsible for the execution of programmed cell death (apoptosis) and must undergo proteolytic activation, in response to apoptotic stimuli, to function. The mechanism of initiator caspase activation has been generalized by the induced proximity model, which is thought to drive dimerization-mediated activation of caspases. The initiator caspase, caspase-9, exists predominantly as a monomer in solution. To examine the induced proximity model, we engineered a constitutively dimeric caspase-9 by relieving steric hindrance at the dimer interface. Crystal structure of the engineered caspase-9 closely resembles that of the wild-type (WT) caspase-9, including all relevant structural details and the asymmetric nature of two monomers. Compared to the WT caspase-9, this engineered dimer exhibits a higher level of catalytic activity in vitro and induces more efficient cell death when expressed. However, the catalytic activity of the dimeric caspase-9 is only a small fraction of that for the Apaf-1-activated caspase-9. Furthermore, in contrast to the WT caspase-9, the activity of the dimeric caspase-9 can no longer be significantly enhanced in an Apaf-1-dependent manner. These findings suggest that dimerization of caspase-9 may be qualitatively different from its activation by Apaf-1, and in conjunction with other evidence, posit an induced conformation model for the activation of initiator caspases.


Subject(s)
Caspases/genetics , Caspases/metabolism , Genetic Engineering , Caspase 9 , Caspases/chemistry , Catalysis , Cloning, Molecular , Crystallography, X-Ray , Dimerization , Enzyme Activation , Escherichia coli/genetics , Models, Biological , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
16.
J Biol Chem ; 279(44): 45844-54, 2004 Oct 29.
Article in English | MEDLINE | ID: mdl-15294909

ABSTRACT

Presenilin mutations are responsible for most cases of autosomal dominant inherited forms of early onset Alzheimer disease. Presenilins play an important role in amyloid beta-precursor processing, NOTCH receptor signaling, and apoptosis. However, the molecular mechanisms by which presenilins regulate apoptosis are not fully understood. Here, we report that presenilin-1 (PS1) regulates the proteolytic activity of the serine protease Omi/HtrA2 through direct interaction with its regulatory PDZ domain. We show that a peptide corresponding to the cytoplasmic C-terminal tail of PS1 dramatically increases the proteolytic activity of Omi/HtrA2 toward the inhibitor of apoptosis proteins and beta-casein and induces cell death in an Omi/HtrA2-dependent manner. Consistent with these results, ectopic expression of full-length PS1, but not PS1 lacking the C-terminal PDZ binding motif, potentiated Omi/HtrA2-induced cell death. Our results suggest that the C terminus of PS1 is an activation peptide ligand for the PDZ domain of Omi/HtrA2 and may regulate the protease activity of Omi/HtrA2 after its release from the mitochondria during apoptosis. This mechanism of Omi/HtrA2 activation is similar to the mechanism of activation of the related bacterial DegS protease by the outer-membrane porins.


Subject(s)
Membrane Proteins/physiology , Serine Endopeptidases/metabolism , Amino Acid Sequence , Apoptosis , Bacterial Proteins/metabolism , Enzyme Activation , HeLa Cells , High-Temperature Requirement A Serine Peptidase 2 , Humans , Membrane Proteins/chemistry , Mitochondrial Proteins , Molecular Sequence Data , Presenilin-1 , Presenilin-2
17.
Nature ; 429(6987): 75-9, 2004 May 06.
Article in English | MEDLINE | ID: mdl-15129283

ABSTRACT

Caspases mediate essential key proteolytic events in inflammatory cascades and the apoptotic cell death pathway. Human caspases functionally segregate into two distinct subfamilies: those involved in cytokine maturation (caspase-1, -4 and -5) and those involved in cellular apoptosis (caspase-2, -3, -6, -7, -8, -9 and -10). Although caspase-12 is phylogenetically related to the cytokine maturation caspases, in mice it has been proposed as a mediator of apoptosis induced by endoplasmic reticulum stress including amyloid-beta cytotoxicity, suggesting that it might contribute to the pathogenesis of Alzheimer's disease. Here we show that a single nucleotide polymorphism in caspase-12 in humans results in the synthesis of either a truncated protein (Csp12-S) or a full-length caspase proenzyme (Csp12-L). The read-through single nucleotide polymorphism encoding Csp12-L is confined to populations of African descent and confers hypo-responsiveness to lipopolysaccharide-stimulated cytokine production in ex vivo whole blood, but has no significant effect on apoptotic sensitivity. In a preliminary study, we find that the frequency of the Csp12-L allele is increased in African American individuals with severe sepsis. Thus, Csp12-L attenuates the inflammatory and innate immune response to endotoxins and in doing so may constitute a risk factor for developing sepsis.


Subject(s)
Caspases/genetics , Lipopolysaccharides/pharmacology , Polymorphism, Single Nucleotide/genetics , Sepsis/genetics , Africa/ethnology , Black or African American/genetics , Alzheimer Disease/genetics , Animals , Apoptosis/drug effects , Base Sequence , Case-Control Studies , Caspase 12 , Caspases/chemistry , Concanavalin A/pharmacology , Cytokines/blood , Endoplasmic Reticulum/metabolism , Gene Frequency , Genetic Predisposition to Disease/genetics , Genotype , Humans , Inflammation/genetics , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Primates/genetics
18.
Oncogene ; 23(13): 2339-46, 2004 Mar 25.
Article in English | MEDLINE | ID: mdl-14767475

ABSTRACT

Glioblastoma multiforme, the most common brain tumor, typically exhibits markedly increased angiogenesis, which is crucial for tumor growth and invasion. Antiangiogenic strategies based on disruption of the tumor microvasculature have proven effective for the treatment of experimental brain tumors. Here, we have overexpressed human caspase-9 by stable transfection in the SNB19 glioblastoma cell line, which normally expresses low levels of caspase-9. Our studies revealed that overexpression of caspase-9 coupled with radiation has a synergistic effect on the inhibition of glioma invasion as demonstrated by Matrigel assay (> 65%). Furthermore, sense caspase stable clones cocultured with fetal rat brain aggregates along with radiation showed complete inhibition as compared to the parental and vector controls. During in vitro angiogenesis, SNB19 cells cocultured with human microvascular endothelial cells (HMEC) showed vascular network formation after 48-72 h. In contrast, these capillary-like structures were inhibited when HMEC cells were cocultured with sense caspase stable SNB19 cells. This effect was further enhanced by radiation (5 Gy). Signaling mechanisms revealed that apoptosis is induced by cleavage of caspase-9 by radiation, loss of mitochondrial membrane potential and activation of caspase-3. These results demonstrate that activation of caspase-9 disrupts glioma cell invasion and angiogenesis in vitro. Hence, overexpression of proapoptotic molecules such as caspase-9 may be an important determinant of the therapeutic effect of radiation in cancer therapy.


Subject(s)
Caspases/radiation effects , Glioma/radiotherapy , Neoplasm Invasiveness , Neovascularization, Pathologic/radiotherapy , Animals , Apoptosis/radiation effects , Capillaries/enzymology , Capillaries/growth & development , Caspase 9 , Caspases/metabolism , Cytochromes c/biosynthesis , Cytochromes c/genetics , Humans , Membrane Potentials/physiology , Mice , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics
19.
Nature ; 425(6959): 721-7, 2003 Oct 16.
Article in English | MEDLINE | ID: mdl-14534547

ABSTRACT

The mouse mutant mnd2 (motor neuron degeneration 2) exhibits muscle wasting, neurodegeneration, involution of the spleen and thymus, and death by 40 days of age. Degeneration of striatal neurons, with astrogliosis and microglia activation, begins at around 3 weeks of age, and other neurons are affected at later stages. Here we have identified the mnd2 mutation as the missense mutation Ser276Cys in the protease domain of the nuclear-encoded mitochondrial serine protease Omi (also known as HtrA2 or Prss25). Protease activity of Omi is greatly reduced in tissues of mnd2 mice but is restored in mice rescued by a bacterial artificial chromosome transgene containing the wild-type Omi gene. Deletion of the PDZ domain partially restores protease activity to the inactive recombinant Omi protein carrying the Ser276Cys mutation, suggesting that the mutation impairs substrate access or binding to the active site pocket. Loss of Omi protease activity increases the susceptibility of mitochondria to induction of the permeability transition, and increases the sensitivity of mouse embryonic fibroblasts to stress-induced cell death. The neurodegeneration and juvenile lethality in mnd2 mice result from this defect in mitochondrial Omi protease.


Subject(s)
Mitochondria/enzymology , Mutation, Missense/genetics , Neuromuscular Diseases/enzymology , Neuromuscular Diseases/genetics , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Amino Acid Sequence , Animals , Binding Sites , Calcium/metabolism , Caseins/genetics , Caseins/metabolism , Cell Death , Cells, Cultured , Chromosome Mapping , Crosses, Genetic , Female , High-Temperature Requirement A Serine Peptidase 2 , Homozygote , Humans , Male , Mice , Mice, Neurologic Mutants , Mice, Transgenic , Mitochondrial Proteins , Molecular Sequence Data , Neuromuscular Diseases/metabolism , Serine Endopeptidases/chemistry , Structure-Activity Relationship
20.
J Biol Chem ; 278(40): 38699-706, 2003 Oct 03.
Article in English | MEDLINE | ID: mdl-12865429

ABSTRACT

Smac/Diablo and HtrA2/Omi are inhibitors of apoptosis (IAP)-binding proteins released from the mitochondria of human cells during apoptosis and regulate apoptosis by liberating caspases from IAP inhibition. Here we describe the identification of a proteolytically processed isoform of the polypeptide chain-releasing factor GSPT1/eRF3 protein, which functions in translation, as a new IAP-binding protein. In common with other IAP-binding proteins, the processed GSPT1 protein harbors a conserved N-terminal IAP-binding motif (AKPF). Additionally, processed GSPT1 interacts biochemically with IAPs and could promote caspase activation, IAP ubiquitination and apoptosis. The IAP-binding motif of the processed GSPT1 is absolutely required for these activities. Our findings are consistent with a model whereby processing of GSPT1 into the IAP-binding isoform could potentiate apoptosis by liberating caspases from IAP inhibition, or target IAPs and the processed GSPT1 for proteasome-mediated degradation.


Subject(s)
Peptide Termination Factors/chemistry , Peptide Termination Factors/physiology , Amino Acid Motifs , Amino Acid Sequence , Apoptosis , Blotting, Western , Caspases/metabolism , Cell Line , Cloning, Molecular , Cysteine Endopeptidases/metabolism , Cytochrome c Group/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Endoplasmic Reticulum/metabolism , Enzyme Activation , Epitopes/chemistry , Glutathione Transferase/metabolism , Humans , Microscopy, Confocal , Mitochondria/metabolism , Molecular Sequence Data , Multienzyme Complexes/metabolism , Proteasome Endopeptidase Complex , Protein Binding , Protein Biosynthesis , Protein Isoforms , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Subcellular Fractions , Time Factors , Transfection , Tumor Cells, Cultured , Ubiquitin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...