Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Gene Regul Mech ; 1867(2): 195029, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38642633

ABSTRACT

The perception that the nucleoli are merely the organelles where ribosome biogenesis occurs is challenged. Only around 30 % of nucleolar proteins are solely involved in producing ribosomes. Instead, the nucleolus plays a critical role in controlling protein trafficking during stress and, according to its dynamic nature, undergoes continuous protein exchange with nucleoplasm under various cellular stressors. Hence, the concept of nucleolar stress has evolved as cellular insults that disrupt the structure and function of the nucleolus. Considering the emerging role of this organelle in DNA repair and the fact that rDNAs are the most fragile genomic loci, therapies targeting the nucleoli are increasingly being developed. Besides, drugs that target ribosome synthesis and induce nucleolar stress can be used in cancer therapy. In contrast, agents that regulate nucleolar activity may be a potential treatment for neurodegeneration caused by abnormal protein accumulation in the nucleolus. Here, I explore the roles of nucleoli beyond their ribosomal functions, highlighting the factors triggering nucleolar stress and their impact on genomic stability.


Subject(s)
Cell Nucleolus , Genomic Instability , Stress, Physiological , Cell Nucleolus/metabolism , Humans , Ribosomes/metabolism , Animals , DNA Repair , Neoplasms/metabolism , Neoplasms/genetics , Neoplasms/pathology , DNA, Ribosomal/metabolism , DNA, Ribosomal/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics
2.
Int J Mol Sci ; 24(17)2023 Sep 04.
Article in English | MEDLINE | ID: mdl-37686461

ABSTRACT

The origin of eukaryotic organisms involved the integration of mitochondria into the ancestor cell, with a massive gene transfer from the original proteobacterium to the host nucleus. Thus, mitochondrial performance relies on a mosaic of nuclear gene products from a variety of genomes. The concerted regulation of their synthesis is necessary for metabolic housekeeping and stress response. This governance involves crosstalk between mitochondrial, cytoplasmic, and nuclear factors. While anterograde and retrograde regulation preserve mitochondrial homeostasis, the mitochondria can modulate a wide set of nuclear genes in response to an extensive variety of conditions, whose response mechanisms often merge. In this review, we summarise how mitochondrial metabolites and proteins-encoded either in the nucleus or in the organelle-target the cell nucleus and exert different actions modulating gene expression and the chromatin state, or even causing DNA fragmentation in response to common stress conditions, such as hypoxia, oxidative stress, unfolded protein stress, and DNA damage.


Subject(s)
Cell Nucleus , Mitochondria , Mitochondria/genetics , Nuclear Proteins , Chromatin , Cytoplasm
3.
Nat Struct Mol Biol ; 29(10): 1024-1036, 2022 10.
Article in English | MEDLINE | ID: mdl-36220893

ABSTRACT

The regular functioning of the nucleolus and nucleus-mitochondria crosstalk are considered unrelated processes, yet cytochrome c (Cc) migrates to the nucleus and even the nucleolus under stress conditions. Nucleolar liquid-liquid phase separation usually serves the cell as a fast, smart mechanism to control the spatial localization and trafficking of nuclear proteins. Actually, the alternative reading frame (ARF), a tumor suppressor protein sequestered by nucleophosmin (NPM) in the nucleoli, is shifted out from NPM upon DNA damage. DNA damage also triggers early translocation of respiratory Cc to nucleus before cytoplasmic caspase activation. Here, we show that Cc can bind to nucleolar NPM by triggering an extended-to-compact conformational change, driving ARF release. Such a NPM-Cc nucleolar interaction can be extended to a general mechanism for DNA damage in which the lysine-rich regions of Cc-rather than the canonical, arginine-rich stretches of membrane-less organelle components-controls the trafficking and availability of nucleolar proteins.


Subject(s)
Cytochromes c , Nucleophosmin , Arginine , Caspases , Lysine , Mitochondria/metabolism , Nuclear Proteins/metabolism , Tumor Suppressor Proteins
4.
Comput Struct Biotechnol J ; 20: 3695-3707, 2022.
Article in English | MEDLINE | ID: mdl-35891793

ABSTRACT

Intrinsic protein flexibility is of overwhelming relevance for intermolecular recognition and adaptability of highly dynamic ensemble of complexes, and the phenomenon is essential for the understanding of numerous biological processes. These conformational ensembles-encounter complexes-lack a unique organization, which prevents the determination of well-defined high resolution structures. This is the case for complexes involving the oncoprotein SET/template-activating factor-Iß (SET/TAF-Iß), a histone chaperone whose functions and interactions are significantly affected by its intrinsic structural plasticity. Besides its role in chromatin remodeling, SET/TAF-Iß is an inhibitor of protein phosphatase 2A (PP2A), which is a key phosphatase counteracting transcription and signaling events controlling the activity of DNA damage response (DDR) mediators. During DDR, SET/TAF-Iß is sequestered by cytochrome c (Cc) upon migration of the hemeprotein from mitochondria to the cell nucleus. Here, we report that the nuclear SET/TAF-Iß:Cc polyconformational ensemble is able to activate PP2A. In particular, the N-end folded, globular region of SET/TAF-Iß (a.k.a. SET/TAF-Iß ΔC)-which exhibits an unexpected, intrinsically highly dynamic behavior-is sufficient to be recognized by Cc in a diffuse encounter manner. Cc-mediated blocking of PP2A inhibition is deciphered using an integrated structural and computational approach, combining small-angle X-ray scattering, electron paramagnetic resonance, nuclear magnetic resonance, calorimetry and molecular dynamics simulations.

5.
FEBS Open Bio ; 11(9): 2418-2440, 2021 09.
Article in English | MEDLINE | ID: mdl-33938164

ABSTRACT

Despite mitochondria being key for the control of cell homeostasis and fate, their role in DNA damage response is usually just regarded as an apoptotic trigger. However, growing evidence points to mitochondrial factors modulating nuclear functions. Remarkably, after DNA damage, cytochrome c (Cc) interacts in the cell nucleus with a variety of well-known histone chaperones, whose activity is competitively inhibited by the haem protein. As nuclear Cc inhibits the nucleosome assembly/disassembly activity of histone chaperones, it might indeed affect chromatin dynamics and histone deposition on DNA. Several histone chaperones actually interact with Cc Lys residues through their acidic regions, which are also involved in heterotypic interactions leading to liquid-liquid phase transitions responsible for the assembly of nuclear condensates, including heterochromatin. This relies on dynamic histone-DNA interactions that can be modulated by acetylation of specific histone Lys residues. Thus, Cc may have a major regulatory role in DNA repair by fine-tuning nucleosome assembly activity and likely nuclear condensate formation.


Subject(s)
Biomolecular Condensates/metabolism , Cell Nucleus/metabolism , Cytochromes c/metabolism , Histone Chaperones/metabolism , Mitochondria/metabolism , Animals , Cell Nucleus/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly , Cytochromes c/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group C/metabolism , Histones/metabolism , Humans , Mitochondria/genetics
6.
Redox Biol ; 43: 101967, 2021 07.
Article in English | MEDLINE | ID: mdl-33882408

ABSTRACT

Repair of injured DNA relies on nucleosome dismantling by histone chaperones and de-phosphorylation events carried out by Protein Phosphatase 2A (PP2A). Typical histone chaperones are the Acidic leucine-rich Nuclear Phosphoprotein 32 family (ANP32) members, e.g. ANP32A, which is also a well-known PP2A inhibitor (a.k.a. I1PP2A). Here we report the novel interaction between the endogenous family member B-so-called ANP32B-and endogenous cytochrome c in cells undergoing camptothecin-induced DNA damage. Soon after DNA lesions but prior to caspase cascade activation, the hemeprotein translocates to the nucleus to target the Low Complexity Acidic Region (LCAR) of ANP32B; in a similar way, our group recently reported that the hemeprotein targets the acidic domain of SET/Template Activating Factor-Iß (SET/TAF-Iß), which is another histone chaperone and PP2A inhibitor (a.k.a. I2PP2A). The nucleosome assembly activity of ANP32B is indeed unaffected by cytochrome c binding. Like ANP32A, ANP32B inhibits PP2A activity and is thus herein referred to as I3PP2A. Our data demonstrates that ANP32B-dependent inhibition of PP2A is regulated by respiratory cytochrome c, which induces long-distance allosteric changes in the structured N-terminal domain of ANP32B upon binding to the C-terminal LCAR. In agreement with the reported role of PP2A in the DNA damage response, we propose a model wherein cytochrome c is translocated from the mitochondria into the nucleus upon DNA damage to modulate PP2A activity via its interaction with ANP32B.


Subject(s)
Cytochromes c , Histone Chaperones , Cell Nucleus , DNA Damage , Protein Phosphatase 2
7.
Plant J ; 106(1): 74-85, 2021 04.
Article in English | MEDLINE | ID: mdl-33354856

ABSTRACT

Programmed cell death (PCD) is crucial for development and homeostasis of all multicellular organisms. In human cells, the double role of extra-mitochondrial cytochrome c in triggering apoptosis and inhibiting survival pathways is well reported. In plants, however, the specific role of cytochrome c upon release from the mitochondria remains in part veiled yet death stimuli do trigger cytochrome c translocation as well. Here, we identify an Arabidopsis thaliana 14-3-3ι isoform as a cytosolic cytochrome c target and inhibitor of caspase-like activity. This finding establishes the 14-3-3ι protein as a relevant factor at the onset of plant H2 O2 -induced PCD. The in vivo and in vitro studies herein reported reveal that the interaction between cytochrome c and 14-3-3ι exhibits noticeable similarities with the complex formed by their human orthologues. Further analysis of the heterologous complexes between human and plant cytochrome c with plant 14-3-3ι and human 14-3-3ε isoforms corroborated common features. These results suggest that cytochrome c blocks p14-3-3ι so as to inhibit caspase-like proteases, which in turn promote cell death upon H2 O2 treatment. Besides establishing common biochemical features between human and plant PCD, this work sheds light onto the signaling networks of plant cell death.


Subject(s)
14-3-3 Proteins/metabolism , Apoptosis/drug effects , Arabidopsis Proteins/metabolism , Arabidopsis/drug effects , Arabidopsis/metabolism , Cytochromes c/metabolism , Cytochromes c/pharmacology , Hydrogen Peroxide
8.
Comput Struct Biotechnol J ; 18: 1852-1863, 2020.
Article in English | MEDLINE | ID: mdl-32728408

ABSTRACT

Post-translational modifications of proteins expand their functional diversity, regulating the response of cells to a variety of stimuli. Among these modifications, phosphorylation is the most ubiquitous and plays a prominent role in cell signaling. The addition of a phosphate often affects the function of a protein by altering its structure and dynamics. However, these alterations are often difficult to study and the functional and structural implications remain unresolved. New approaches are emerging to overcome common obstacles related to the production and manipulation of these samples. Here, we summarize the available methods for phosphoprotein purification and phosphomimetic engineering, highlighting the advantages and disadvantages of each. We propose a general workflow for protein phosphorylation analysis combining computational and biochemical approaches, building on recent advances that enable user-friendly and easy-to-access Molecular Dynamics simulations. We hope this innovative workflow will inform the best experimental approach to explore such post-translational modifications. We have applied this workflow to two different human protein models: the hemeprotein cytochrome c and the RNA binding protein HuR. Our results illustrate the usefulness of Molecular Dynamics as a decision-making tool to design the most appropriate phosphomimetic variant.

9.
FEBS Lett ; 593(22): 3101-3119, 2019 11.
Article in English | MEDLINE | ID: mdl-31663111

ABSTRACT

Cytochrome c (Cc) is a protein that functions as an electron carrier in the mitochondrial respiratory chain. However, Cc has moonlighting roles outside mitochondria driving the transition of apoptotic cells from life to death. When living cells are damaged, Cc escapes its natural mitochondrial environment and, once in the cytosol, it binds other proteins to form a complex named the apoptosome-a platform that triggers caspase activation and further leads to controlled cell dismantlement. Early released Cc also binds to inositol 1,4,5-triphosphate receptors on the ER membrane, which stimulates further massive Cc release from mitochondria. Besides the well-characterized binding proteins contributing to the proapoptotic functions of Cc, many novel protein targets have been recently described. Among them, histone chaperones were identified as key partners of Cc following DNA breaks, indicating that Cc might modulate chromatin dynamics through competitive binding to histone chaperones. In this article, we review the ample set of recently discovered antiapoptotic proteins-involved in DNA damage, transcription, and energetic metabolism-reported to interact with Cc in the cytoplasm and even the nucleus upon DNA breaks.


Subject(s)
Cell Nucleus/metabolism , Cytochromes c/metabolism , Cytoplasm/metabolism , Chromatin Assembly and Disassembly , Histone Chaperones/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mitochondria/metabolism
10.
Proc Natl Acad Sci U S A ; 115(31): 7955-7960, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30018060

ABSTRACT

Respiratory cytochrome c has been found to be phosphorylated at tyrosine 97 in the postischemic brain upon neuroprotective insulin treatment, but how such posttranslational modification affects mitochondrial metabolism is unclear. Here, we report the structural features and functional behavior of a phosphomimetic cytochrome c mutant, which was generated by site-specific incorporation at position 97 of p-carboxymethyl-l-phenylalanine using the evolved tRNA synthetase method. We found that the point mutation does not alter the overall folding and heme environment of cytochrome c, but significantly affects the entire oxidative phosphorylation process. In fact, the electron donation rate of the mutant heme protein to cytochrome c oxidase, or complex IV, within respiratory supercomplexes was higher than that of the wild-type species, in agreement with the observed decrease in reactive oxygen species production. Direct contact of cytochrome c with the respiratory supercomplex factor HIGD1A (hypoxia-inducible domain family member 1A) is reported here, with the mutant heme protein exhibiting a lower affinity than the wild-type species. Interestingly, phosphomimetic cytochrome c also exhibited a lower caspase-3 activation activity. Altogether, these findings yield a better understanding of the molecular basis for mitochondrial metabolism in acute diseases, such as brain ischemia, and thus could allow the use of phosphomimetic cytochrome c as a neuroprotector with therapeutic applications.


Subject(s)
Cytochromes c/metabolism , Electron Transport Complex IV/metabolism , Mitochondria/enzymology , Mutation , Oxidative Stress , Animals , Caspase 3/genetics , Caspase 3/metabolism , Cattle , Cell Line , Cytochromes c/genetics , Humans , Intracellular Signaling Peptides and Proteins , Mitochondria/genetics , Mitochondrial Proteins , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phosphorylation , Rabbits
11.
Cell Death Dis ; 9(3): 365, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29511177

ABSTRACT

Apoptosis is a highly regulated form of programmed cell death, essential to the development and homeostasis of multicellular organisms. Cytochrome c is a central figure in the activation of the apoptotic intrinsic pathway, thereby activating the caspase cascade through its interaction with Apaf-1. Our recent studies have revealed 14-3-3ε (a direct inhibitor of Apaf-1) as a cytosolic cytochrome c target. Here we explore the cytochrome c / 14-3-3ε interaction and show the ability of cytochrome c to block 14-3-3ε-mediated Apaf-1 inhibition, thereby unveiling a novel function for cytochrome c as an indirect activator of caspase-9/3. We have used calorimetry, NMR spectroscopy, site mutagenesis and computational calculations to provide an insight into the structural features of the cytochrome c / 14-3-3ε complex. Overall, these findings suggest an additional cytochrome c-mediated mechanism to modulate apoptosome formation, shedding light onto the rigorous apoptotic regulation network.


Subject(s)
14-3-3 Proteins/metabolism , Apoptotic Protease-Activating Factor 1/metabolism , Caspase 3/metabolism , Caspase 9/metabolism , Cytochromes c/metabolism , 14-3-3 Proteins/chemistry , 14-3-3 Proteins/genetics , Amino Acid Motifs , Apoptotic Protease-Activating Factor 1/genetics , Caspase 3/genetics , Caspase 9/genetics , Cytosol/enzymology , Cytosol/metabolism , Enzyme Activation , Humans , Protein Binding
12.
Proc Natl Acad Sci U S A ; 114(15): E3041-E3050, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28348229

ABSTRACT

Regulation of mitochondrial activity allows cells to adapt to changing conditions and to control oxidative stress, and its dysfunction can lead to hypoxia-dependent pathologies such as ischemia and cancer. Although cytochrome c phosphorylation-in particular, at tyrosine 48-is a key modulator of mitochondrial signaling, its action and molecular basis remain unknown. Here we mimic phosphorylation of cytochrome c by replacing tyrosine 48 with p-carboxy-methyl-l-phenylalanine (pCMF). The NMR structure of the resulting mutant reveals significant conformational shifts and enhanced dynamics around pCMF that could explain changes observed in its functionality: The phosphomimetic mutation impairs cytochrome c diffusion between respiratory complexes, enhances hemeprotein peroxidase and reactive oxygen species scavenging activities, and hinders caspase-dependent apoptosis. Our findings provide a framework to further investigate the modulation of mitochondrial activity by phosphorylated cytochrome c and to develop novel therapeutic approaches based on its prosurvival effects.


Subject(s)
Cytochromes c/metabolism , Mitochondria/pathology , Oxidative Stress , Reactive Oxygen Species/metabolism , Tyrosine/chemistry , Cytochromes c/chemistry , Cytochromes c/genetics , Humans , Magnetic Resonance Spectroscopy , Mitochondria/metabolism , Mutation , Peroxidases/metabolism , Phenylalanine/analogs & derivatives , Phenylalanine/chemistry , Phenylalanine/metabolism , Phosphorylation , Protein Conformation , Signal Transduction , Tyrosine/genetics , Tyrosine/metabolism
13.
Nucleic Acids Res ; 45(4): 2150-2165, 2017 02 28.
Article in English | MEDLINE | ID: mdl-27924001

ABSTRACT

Higher-order plants and mammals use similar mechanisms to repair and tolerate oxidative DNA damage. Most studies on the DNA repair process have focused on yeast and mammals, in which histone chaperone-mediated nucleosome disassembly/reassembly is essential for DNA to be accessible to repair machinery. However, little is known about the specific role and modulation of histone chaperones in the context of DNA damage in plants. Here, the histone chaperone NRP1, which is closely related to human SET/TAF-Iß, was found to exhibit nucleosome assembly activity in vitro and to accumulate in the chromatin of Arabidopsis thaliana after DNA breaks. In addition, this work establishes that NRP1 binds to cytochrome c, thereby preventing the former from binding to histones. Since NRP1 interacts with cytochrome c at its earmuff domain, that is, its histone-binding domain, cytochrome c thus competes with core histones and hampers the activity of NRP1 as a histone chaperone. Altogether, the results obtained indicate that the underlying molecular mechanisms in nucleosome disassembly/reassembly are highly conserved throughout evolution, as inferred from the similar inhibition of plant NRP1 and human SET/TAF-Iß by cytochrome c during DNA damage response.


Subject(s)
Arabidopsis Proteins/metabolism , Cytochromes c/metabolism , Histone Chaperones/metabolism , Arabidopsis , Arabidopsis Proteins/chemistry , Cells, Cultured , Chromatin/genetics , Chromatin/metabolism , Cytochromes c/chemistry , DNA Damage , DNA-Binding Proteins , Histone Chaperones/chemistry , Histones/metabolism , Humans , Models, Molecular , Nucleosomes/metabolism , Plant Cells , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Thermodynamics , Transcription Factors/metabolism
14.
Proc Natl Acad Sci U S A ; 112(32): 9908-13, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26216969

ABSTRACT

Chromatin is pivotal for regulation of the DNA damage process insofar as it influences access to DNA and serves as a DNA repair docking site. Recent works identify histone chaperones as key regulators of damaged chromatin's transcriptional activity. However, understanding how chaperones are modulated during DNA damage response is still challenging. This study reveals that the histone chaperone SET/TAF-Iß interacts with cytochrome c following DNA damage. Specifically, cytochrome c is shown to be translocated into cell nuclei upon induction of DNA damage, but not upon stimulation of the death receptor or stress-induced pathways. Cytochrome c was found to competitively hinder binding of SET/TAF-Iß to core histones, thereby locking its histone-binding domains and inhibiting its nucleosome assembly activity. In addition, we have used NMR spectroscopy, calorimetry, mutagenesis, and molecular docking to provide an insight into the structural features of the formation of the complex between cytochrome c and SET/TAF-Iß. Overall, these findings establish a framework for understanding the molecular basis of cytochrome c-mediated blocking of SET/TAF-Iß, which subsequently may facilitate the development of new drugs to silence the oncogenic effect of SET/TAF-Iß's histone chaperone activity.


Subject(s)
Cytochromes c/chemistry , Cytochromes c/metabolism , Histone Chaperones/antagonists & inhibitors , Histone Chaperones/chemistry , Histones/metabolism , Molecular Chaperones/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Transcription Factors/chemistry , Animals , Binding, Competitive/drug effects , Camptothecin/pharmacology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , DNA Damage , DNA-Binding Proteins , HeLa Cells , Histone Chaperones/metabolism , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Protein Binding/drug effects , Protein Structure, Tertiary , Protein Transport/drug effects , Structure-Activity Relationship , Transcription Factors/metabolism , Xenopus
15.
FEBS Lett ; 589(4): 476-83, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25595453

ABSTRACT

The transient interactions of respiratory cytochrome c with complexes III and IV is herein investigated by using heterologous proteins, namely human cytochrome c, the soluble domain of plant cytochrome c1 and bovine cytochrome c oxidase. The binding molecular mechanisms of the resulting cross-complexes have been analyzed by Nuclear Magnetic Resonance and Isothermal Titration Calorimetry. Our data reveal that the two cytochrome c-involving adducts possess a 2:1 stoichiometry - that is, two cytochrome c molecules per adduct - at low ionic strength. We conclude that such extra binding sites at the surfaces of complexes III and IV can facilitate the turnover and sliding of cytochrome c molecules and, therefore, the electron transfer within respiratory supercomplexes.


Subject(s)
Cytochromes c/chemistry , Electron Transport Complex III/chemistry , Electron Transport Complex IV/chemistry , Animals , Cattle , Humans , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Osmolar Concentration , Protein Binding , Protein Structure, Quaternary , Solutions
16.
Biochim Biophys Acta ; 1837(10): 1717-29, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25091281

ABSTRACT

In plants, channeling of cytochrome c molecules between complexes III and IV has been purported to shuttle electrons within the supercomplexes instead of carrying electrons by random diffusion across the intermembrane bulk phase. However, the mode plant cytochrome c behaves inside a supercomplex such as the respirasome, formed by complexes I, III and IV, remains obscure from a structural point of view. Here, we report ab-initio Brownian dynamics calculations and nuclear magnetic resonance-driven docking computations showing two binding sites for plant cytochrome c at the head soluble domain of plant cytochrome c1, namely a non-productive (or distal) site with a long heme-to-heme distance and a functional (or proximal) site with the two heme groups close enough as to allow electron transfer. As inferred from isothermal titration calorimetry experiments, the two binding sites exhibit different equilibrium dissociation constants, for both reduced and oxidized species, that are all within the micromolar range, thus revealing the transient nature of such a respiratory complex. Although the docking of cytochrome c at the distal site occurs at the interface between cytochrome c1 and the Rieske subunit, it is fully compatible with the complex III structure. In our model, the extra distal site in complex III could indeed facilitate the functional cytochrome c channeling towards complex IV by building a "floating boat bridge" of cytochrome c molecules (between complexes III and IV) in plant respirasome.


Subject(s)
Cytochromes c1/metabolism , Plants/enzymology , Base Sequence , Binding Sites , Cytochromes c1/genetics , DNA Primers , Magnetic Resonance Spectroscopy , Molecular Docking Simulation , Mutagenesis, Site-Directed , Polymerase Chain Reaction , Ultracentrifugation
17.
Mol Cell Proteomics ; 13(6): 1439-56, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24643968

ABSTRACT

Since the first description of apoptosis four decades ago, great efforts have been made to elucidate, both in vivo and in vitro, the molecular mechanisms involved in its regulation. Although the role of cytochrome c during apoptosis is well established, relatively little is known about its participation in signaling pathways in vivo due to its essential role during respiration. To obtain a better understanding of the role of cytochrome c in the onset of apoptosis, we used a proteomic approach based on affinity chromatography with cytochrome c as bait in this study. In this approach, novel cytochrome c interaction partners were identified whose in vivo interaction and cellular localization were facilitated through bimolecular fluorescence complementation. Modeling of the complex interface between cytochrome c and its counterparts indicated the involvement of the surface surrounding the heme crevice of cytochrome c, in agreement with the vast majority of known redox adducts of cytochrome c. However, in contrast to the high turnover rate of the mitochondrial cytochrome c redox adducts, those occurring under apoptosis led to the formation of stable nucleo-cytoplasmic ensembles, as inferred mainly from surface plasmon resonance and nuclear magnetic resonance measurements, which permitted us to corroborate the formation of such complexes in vitro. The results obtained suggest that human cytochrome c interacts with pro-survival, anti-apoptotic proteins following its release into the cytoplasm. Thus, cytochrome c may interfere with cell survival pathways and unlock apoptosis in order to prevent the spatial and temporal coexistence of antagonist signals.


Subject(s)
Apoptosis/genetics , Cytochromes c/biosynthesis , Cytochromes c/chemistry , Proteomics , Caspase 3/metabolism , Cell Survival/genetics , Crystallography, X-Ray , Cytochromes c/metabolism , Humans , Mitochondria/genetics , Mitochondria/metabolism , Signal Transduction/genetics
18.
Mol Cell Proteomics ; 12(12): 3666-76, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24019145

ABSTRACT

Programmed cell death is an event displayed by many different organisms along the evolutionary scale. In plants, programmed cell death is necessary for development and the hypersensitive response to stress or pathogenic infection. A common feature in programmed cell death across organisms is the translocation of cytochrome c from mitochondria to the cytosol. To better understand the role of cytochrome c in the onset of programmed cell death in plants, a proteomic approach was developed based on affinity chromatography and using Arabidopsis thaliana cytochrome c as bait. Using this approach, ten putative new cytochrome c partners were identified. Of these putative partners and as indicated by bimolecular fluorescence complementation, nine of them bind the heme protein in plant protoplasts and human cells as a heterologous system. The in vitro interaction between cytochrome c and such soluble cytochrome c-targets was further corroborated using surface plasmon resonance. Taken together, the results obtained in the study indicate that Arabidopsis thaliana cytochrome c interacts with several distinct proteins involved in protein folding, translational regulation, cell death, oxidative stress, DNA damage, energetic metabolism, and mRNA metabolism. Interestingly, some of these novel Arabidopsis thaliana cytochrome c-targets are closely related to those for Homo sapiens cytochrome c (Martínez-Fábregas et al., unpublished). These results indicate that the evolutionarily well-conserved cytosolic cytochrome c, appearing in organisms from plants to mammals, interacts with a wide range of targets on programmed cell death. The data have been deposited to the ProteomeXchange with identifier PXD000280.


Subject(s)
Apoptosis/genetics , Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Cytochromes c/metabolism , Gene Expression Regulation, Plant , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Chromatography, Affinity , Cytochromes c/genetics , Cytosol/chemistry , Cytosol/metabolism , Energy Metabolism , Evolution, Molecular , HEK293 Cells , Humans , Mass Spectrometry , Mitochondria/chemistry , Mitochondria/metabolism , Molecular Sequence Annotation , Oxidative Stress , Protein Binding , Protein Interaction Mapping , Protein Transport , Protoplasts/chemistry , Protoplasts/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Surface Plasmon Resonance
19.
Chemphyschem ; 14(13): 3095-102, 2013 Sep 16.
Article in English | MEDLINE | ID: mdl-23630195

ABSTRACT

Tyrosine nitration is a common post-translational modification affecting protein structure and function. It is based on the addition of a -NO2 group at the ortho position of the phenolic hydroxyl group of tyrosine to yield 3-nitrotyrosine (3-NTyr). Understanding how tyrosine nitration affects the structure and functionality of proteins is of considerable interest, as it is associated with pathogenesis in diseases related to oxidative stress in all living organisms. There are several methods to nitrate tyrosine residues in native proteins. Among them, nitration by the chemical agent peroxynitrite stands out for its biological relevance. Recently, a genetically evolved suppressor tRNA has been developed to provide in vivo incorporation of 3-NTyr into proteins. In this minireview, we discuss the advantages and limitations of these chemical and biological methods and propose a non-damaging method to analyze the configuration and dynamics of nitrotyrosine residues in native proteins by NMR spectroscopy.


Subject(s)
Proteins/chemistry , Tyrosine/analogs & derivatives , Mitochondria/enzymology , Mitochondria/metabolism , Molecular Structure , Nitrates/chemistry , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase/metabolism , Tyrosine/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...