Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Immunol ; 208(7): 1729-1741, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35277420

ABSTRACT

Recent evidence suggests interaction of platelets with dendritic cells (DCs), while the molecular mechanisms mediating this heterotypic cell cross-talk are largely unknown. We evaluated the role of integrin Mac-1 (αMß2, CD11b/CD18) on DCs as a counterreceptor for platelet glycoprotein (GP) Ibα. In a dynamic coincubation model, we observed interaction of human platelets with monocyte-derived DCs, but also that platelet activation induced a sharp increase in heterotypic cell binding. Inhibition of CD11b or GPIbα led to significant reduction of DC adhesion to platelets in vitro independent of GPIIbIIIa, which we confirmed using platelets from Glanzmann thrombasthenia patients and transgenic mouse lines on C57BL/6 background (GPIbα-/-, IL4R-GPIbα-tg, and muMac1 mice). In vivo, inhibition or genetic deletion of CD11b and GPIbα induced a significant reduction of platelet-mediated DC adhesion to the injured arterial wall. Interestingly, only intravascular antiCD11b inhibited DC recruitment, suggesting a dynamic DC-platelet interaction. Indeed, we could show that activated platelets induced CD11b upregulation on Mg2+-preactivated DCs, which was related to protein kinase B (Akt) and dependent on P-selectin and P-selectin glycoprotein ligand 1. Importantly, specific pharmacological targeting of the GPIbα-Mac-1 interaction site blocked DC-platelet interaction in vitro and in vivo. These results demonstrate that cross-talk of platelets with DCs is mediated by GPIbα and Mac-1, which is upregulated on DCs by activated platelets in a P-selectin glycoprotein ligand 1-dependent manner.


Subject(s)
Blood Platelets , CD18 Antigens , Animals , Blood Platelets/physiology , CD18 Antigens/metabolism , Cell Adhesion , Cell Communication , Dendritic Cells/metabolism , Humans , Mice , Mice, Inbred C57BL , Platelet Glycoprotein GPIb-IX Complex/metabolism
2.
Mol Cell Proteomics ; 21(2): 100191, 2022 02.
Article in English | MEDLINE | ID: mdl-34974192

ABSTRACT

Mitophagy, the selective degradation of mitochondria by autophagy, affects defective mitochondria following damage or stress. At the onset of mitophagy, parkin ubiquitylates proteins on the mitochondrial outer membrane. While the role of parkin at the onset of mitophagy is well understood, less is known about its activity during later stages in the process. Here, we used HeLa cells expressing catalytically active or inactive parkin to perform temporal analysis of the proteome, ubiquitylome, and phosphoproteome during 18 h after induction of mitophagy by mitochondrial uncoupler carbonyl cyanide m-chlorophenyl hydrazine. Abundance profiles of proteins downregulated in parkin-dependent manner revealed a stepwise and "outside-in" directed degradation of mitochondrial subcompartments. While ubiquitylation of mitochondrial outer membrane proteins was enriched among early parkin-dependent targets, numerous mitochondrial inner membrane, matrix, and cytosolic proteins were also found ubiquitylated at later stages of mitophagy. Phosphoproteome analysis revealed a possible crosstalk between phosphorylation and ubiquitylation during mitophagy on key parkin targets, such as voltage-dependent anion channel 2.


Subject(s)
Mitophagy , Ubiquitin-Protein Ligases , HeLa Cells , Humans , Phosphorylation , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
3.
iScience ; 23(12): 101797, 2020 Dec 18.
Article in English | MEDLINE | ID: mdl-33299968

ABSTRACT

PINK1 loss-of-function mutations cause early onset Parkinson disease. PINK1-Parkin mediated mitophagy has been well studied, but the relevance of the endogenous process in the brain is debated. Here, the absence of PINK1 in human dopaminergic neurons inhibits ionophore-induced mitophagy and reduces mitochondrial membrane potential. Compensatory, mitochondrial renewal maintains mitochondrial morphology and protects the respiratory chain. This is paralleled by metabolic changes, including inhibition of the TCA cycle enzyme mAconitase, accumulation of NAD+, and metabolite depletion. Loss of PINK1 disrupts dopamine metabolism by critically affecting its synthesis and uptake. The mechanism involves steering of key amino acids toward energy production rather than neurotransmitter metabolism and involves cofactors related to the vitamin B6 salvage pathway identified using unbiased multi-omics approaches. We propose that reduction of mitochondrial membrane potential that cannot be controlled by PINK1 signaling initiates metabolic compensation that has neurometabolic consequences relevant to Parkinson disease.

4.
Exp Cell Res ; 384(2): 111641, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31550441

ABSTRACT

Parkin is an ubiquitin ligase regulating mitochondrial quality control reactions, including the autophagic removal of depolarized mitochondria (mitophagy). Parkin-mediated protein ubiquitinations may be counteracted by deubiquitinating enzymes (DUBs). We conducted a high-content imaging screen of Parkin translocation to depolarized mitochondria after siRNA mediated silencing of each DUB in Parkin overexpressing HeLa cells. Knockdown of the ubiquitin-specific protease USP36 led to delayed Parkin translocation while only slightly disturbing the ubiquitination of mitochondrial proteins, but final autophagic elimination of mitochondria was severely disrupted. The localization of the nucleolar USP36 was not altered during mitophagy. However, the marker for transcriptional active chromatin, histone 2B Lys120 mono-ubiquitination was found reduced in USP36-silenced cells undergoing mitophagy. We observed a reduction of the mRNA and protein levels of Beclin-1 and its associated autophagy-related key regulator ATG14L in USP36 knockdown cells. Importantly, transfection of active ATG14L into USP36-silenced cells significantly restored Parkin-dependent mitophagy. We propose USP36 as regulator for the Parkin-dependent mitophagy at least in part via the Beclin-1-ATG14L pathway.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Autophagy-Related Proteins/genetics , Autophagy/genetics , Beclin-1/genetics , Down-Regulation/genetics , Mitophagy/genetics , Ubiquitin Thiolesterase/genetics , Ubiquitin-Protein Ligases/genetics , Cell Line, Tumor , Gene Knockdown Techniques/methods , HeLa Cells , Humans , Mitochondria/genetics , Mitochondrial Proteins/genetics , Ubiquitin/genetics , Ubiquitin-Specific Proteases/genetics , Ubiquitination/genetics
5.
Nucleic Acids Res ; 45(5): 2797-2808, 2017 03 17.
Article in English | MEDLINE | ID: mdl-27907896

ABSTRACT

Site-directed A-to-I RNA editing is a technology for re-programming genetic information at the RNA-level. We describe here the first design of genetically encodable guideRNAs that enable the re-addressing of human ADAR2 toward specific sites in user-defined mRNA targets. Up to 65% editing yield has been achieved in cell culture for the recoding of a premature Stop codon (UAG) into tryptophan (UIG). In the targeted gene, editing was very specific. We applied the technology to recode a recessive loss-of-function mutation in PINK1 (W437X) in HeLa cells and showed functional rescue of PINK1/Parkin-mediated mitophagy, which is linked to the etiology of Parkinson's disease. In contrast to other editing strategies, this approach requires no artificial protein. Our novel guideRNAs may allow for the development of a platform technology that requires only the administration or expression of a guideRNA to recode genetic information, with high potential for application in biology and medicine.


Subject(s)
Adenosine Deaminase/metabolism , Mitophagy , Point Mutation , Protein Kinases/genetics , RNA Editing , RNA-Binding Proteins/metabolism , Codon, Nonsense , Genome , HEK293 Cells , HeLa Cells , Humans , RNA, Guide, Kinetoplastida/chemistry , RNA, Messenger/metabolism , Transfection
6.
Neurobiol Dis ; 71: 1-13, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25107340

ABSTRACT

Mutations in vacuolar protein sorting 35 (VPS35) have been linked to familial Parkinson's disease (PD). VPS35, a component of the retromer, mediates the retrograde transport of cargo from the endosome to the trans-Golgi network. Here we showed that retromer depletion increases the lysosomal turnover of the mannose 6-phosphate receptor, thereby affecting the trafficking of cathepsin D (CTSD), a lysosome protease involved in α-synuclein (αSYN) degradation. VPS35 knockdown perturbed the maturation step of CTSD in parallel with the accumulation of αSYN in the lysosomes. Furthermore, we found that the knockdown of Drosophila VPS35 not only induced the accumulation of the detergent-insoluble αSYN species in the brain but also exacerbated both locomotor impairments and mild compound eye disorganization and interommatidial bristle loss in flies expressing human αSYN. These findings indicate that the retromer may play a crucial role in αSYN degradation by modulating the maturation of CTSD and might thereby contribute to the pathogenesis of the disease.


Subject(s)
Drosophila Proteins/genetics , Lysosomes/metabolism , Mutation/genetics , Parkinson Disease/genetics , Parkinson Disease/metabolism , Vesicular Transport Proteins/genetics , alpha-Synuclein/metabolism , Animals , Animals, Genetically Modified , Brain/metabolism , Brain/pathology , Cathepsin D/metabolism , Disease Models, Animal , Drosophila , Eye/metabolism , Eye/pathology , Gene Expression Regulation/genetics , HEK293 Cells , Humans , Immunoprecipitation , Locomotion/genetics , Parkinson Disease/pathology , Protein Transport/genetics , RNA Interference/physiology , Subcellular Fractions/metabolism , Subcellular Fractions/ultrastructure
7.
J Cell Sci ; 127(Pt 15): 3280-93, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24906799

ABSTRACT

Depolarized mitochondria are degraded by mitophagy in a process that depends on the Parkinson's disease gene products PINK1 and Parkin. This is accompanied by ubiquitylation of several mitochondrial substrates. The roles of E2 ubiquitin-conjugating enzymes (UBE2) in mitophagy are poorly understood. Here, we investigate a set of UBE2 enzymes that might regulate Parkin-mediated mitophagy. Knockdown of the E2 enzymes UBE2N, UBE2L3 or UBE2D2 and UBE2D3 (UBE2D2/3) significantly reduced autophagic clearance of depolarized mitochondria. However, this did not interfere with mitochondrial PINK1 stabilization and Parkin translocation. UBE2N knockdown prevented specifically K63-linked ubiquitylation at mitochondrial sites. Nevertheless, polyubiquitin and p62 (officially known as SQSTM1) were still found on mitochondria after individual UBE2 knockdown. Knockdown of all of these UBE2s together significantly reduced mitochondrial polyubiquitylation and p62 recruitment. Moreover, reduced ubiquitylation of mitofusins, the mitochondrial import receptor subunits TOM20 and TOM70, the voltage-dependent anion channel protein 1 and Parkin was observed in cells silenced for all of these UBE2s. A version of Parkin with a mutation in the active site (C431S) failed to ubiquitylate these mitochondrial substrates even in the presence of UBE2s. We conclude that UBE2N, UBE2L3 and UBE2D2/3 synergistically contribute to Parkin-mediated mitophagy.


Subject(s)
Mitochondria/physiology , Parkinson Disease/enzymology , Protein Kinases/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , HeLa Cells , Homeostasis , Humans , Membrane Potential, Mitochondrial , Membrane Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Precursor Protein Import Complex Proteins , Mitophagy/genetics , Mutation/genetics , Parkinson Disease/genetics , RNA, Small Interfering/genetics , Receptors, Cell Surface/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics , Voltage-Dependent Anion Channel 1/metabolism
8.
J Biol Chem ; 289(27): 19164-79, 2014 Jul 04.
Article in English | MEDLINE | ID: mdl-24825905

ABSTRACT

Trans-activation element DNA-binding protein of 43 kDa (TDP-43) characterizes insoluble protein aggregates in distinct subtypes of frontotemporal lobar degeneration and amyotrophic lateral sclerosis. TDP-43 mediates many RNA processing steps within distinct protein complexes. Here we identify novel TDP-43 protein interactors found in a yeast two-hybrid screen using an adult human brain cDNA library. We confirmed the TDP-43 interaction of seven hits by co-immunoprecipitation and assessed their co-localization in HEK293E cells. As pathological TDP-43 is ubiquitinated, we focused on the ubiquitin-conjugating enzyme UBE2E3 and the ubiquitin isopeptidase Y (UBPY). When cells were treated with proteasome inhibitor, ubiquitinated and insoluble TDP-43 species accumulated. All three UBE2E family members could enhance the ubiquitination of TDP-43, whereas catalytically inactive UBE2E3(C145S) was much less efficient. Conversely, silencing of UBE2E3 reduced TDP-43 ubiquitination. We examined 15 of the 48 known disease-associated TDP-43 mutants and found that one was excessively ubiquitinated. This strong TDP-43(K263E) ubiquitination was further enhanced by proteasomal inhibition as well as UBE2E3 expression. Conversely, UBE2E3 silencing and expression of UBPY reduced TDP-43(K263E) ubiquitination. Moreover, wild-type but not active site mutant UBPY reduced ubiquitination of TDP-43 C-terminal fragments and of a nuclear import-impaired mutant. In Drosophila melanogaster, UBPY silencing enhanced neurodegenerative TDP-43 phenotypes and the accumulation of insoluble high molecular weight TDP-43 and ubiquitin species. Thus, UBE2E3 and UBPY participate in the regulation of TDP-43 ubiquitination, solubility, and neurodegeneration.


Subject(s)
DNA-Binding Proteins/metabolism , Endopeptidases/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination , Adult , Animals , Brain/metabolism , Drosophila melanogaster/metabolism , Endopeptidases/deficiency , Endosomal Sorting Complexes Required for Transport/deficiency , HEK293 Cells , Humans , Neurotoxins/metabolism , Protein Transport , Two-Hybrid System Techniques , Ubiquitin Thiolesterase/deficiency
9.
J Biol Chem ; 289(26): 18137-51, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24831002

ABSTRACT

α-Synuclein (aS) is a major constituent of Lewy bodies, which are not only a pathological marker for Parkinson disease but also a trigger for neurodegeneration. Cumulative evidence suggests that aS spreads from cell to cell and thereby propagates neurodegeneration to neighboring cells. Recently, Nedd4-1 (neural precursor cell expressed developmentally down-regulated protein 4-1), an E3 ubiquitin ligase, was shown to catalyze the Lys-63-linked polyubiquitination of intracellular aS and thereby facilitate aS degradation by the endolysosomal pathway. Because Nedd4-1 exerts its activity in close proximity to the inner leaflet of the plasma membrane, we speculate that after the internalization of aS the membrane resident aS is preferentially ubiquitinated by Nedd4-1. To clarify the role of Nedd4-1 in aS internalization and endolysosomal sequestration, we generated aS mutants, including ΔPR1(1-119 and 129-140), ΔC(1-119), and ΔPR2(1-119 and 134-140), that lack the proline-rich sequence, a putative Nedd4-1 recognition site. We show that wild type aS, but not ΔPR1, ΔPR2, or ΔC aS, is modified by Nedd4-1 in vitro, acquiring a Lys-63-linked ubiquitin chain. Compared with the mutants lacking the proline-rich sequence, wild type-aS is preferentially internalized and translocated to endosomes. The overexpression of Nedd4-1 increased aS in endosomes, whereas RNAi-mediated silencing of Nedd4-1 decreased endosomal aS. Although aS freely passes through plasma membranes within minutes, a pulse-chase experiment revealed that the overexpression of Nedd4-1 markedly decreased the re-secretion of internalized aS. Together, these findings demonstrate that Nedd4-1-linked Lys-63 ubiquitination specifies the fate of extrinsic and de novo synthesized aS by facilitating their targeting to endosomes.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Endosomes/metabolism , Lysine/metabolism , Ubiquitin-Protein Ligases/metabolism , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , Amino Acid Motifs , Cell Line, Tumor , Endosomal Sorting Complexes Required for Transport/genetics , Endosomes/genetics , Humans , Lysine/genetics , Nedd4 Ubiquitin Protein Ligases , Parkinson Disease/genetics , Parkinson Disease/metabolism , Protein Transport , Ubiquitin-Protein Ligases/genetics , Ubiquitination , alpha-Synuclein/genetics
10.
Autophagy ; 6(7): 871-8, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20798600

ABSTRACT

Mitochondrial dysfunction is an early sign of many neurodegenerative diseases. Very recently, two Parkinson disease (PD) associated genes, PINK1 and Parkin, were shown to mediate the degradation of damaged mitochondria via selective autophagy (mitophagy). PINK1 kinase activity is needed for prompt and efficient Parkin recruitment to impaired mitochondria. PD-associated Parkin mutations interfere with the process of mitophagy at distinct steps. Here we show that whole mitochondria are turned over via macroautophagy. Moreover, disease-associated PINK1 mutations also compromise the selective degradation of depolarized mitochondria. This may be due to the decreased physical binding activity of PD-linked PINK1 mutations to Parkin. Thus, PINK1 mutations abrogate autophagy of impaired mitochondria upstream of Parkin. In addition to compromised PINK1 kinase activity, reduced binding of PINK1 to Parkin leads to failure in Parkin mitochondrial translocation, resulting in the accumulation of damaged mitochondria, which may contribute to disease pathogenesis.


Subject(s)
Autophagy/genetics , Mitochondria , Parkinson Disease/genetics , Parkinson Disease/pathology , Protein Kinases/genetics , Ubiquitin-Protein Ligases/genetics , Animals , HEK293 Cells , HeLa Cells , Humans , Mitochondria/pathology , Mitochondria/physiology , Mutation , Parkinson Disease/physiopathology , Protein Kinases/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ubiquitin-Protein Ligases/metabolism
11.
Nat Cell Biol ; 12(2): 119-31, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20098416

ABSTRACT

Parkinson's disease is the most common neurodegenerative movement disorder. Mutations in PINK1 and PARKIN are the most frequent causes of recessive Parkinson's disease. However, their molecular contribution to pathogenesis remains unclear. Here, we reveal important mechanistic steps of a PINK1/Parkin-directed pathway linking mitochondrial damage, ubiquitylation and autophagy in non-neuronal and neuronal cells. PINK1 kinase activity and its mitochondrial localization sequence are prerequisites to induce translocation of the E3 ligase Parkin to depolarized mitochondria. Subsequently, Parkin mediates the formation of two distinct poly-ubiquitin chains, linked through Lys 63 and Lys 27. In addition, the autophagic adaptor p62/SQSTM1 is recruited to mitochondrial clusters and is essential for the clearance of mitochondria. Strikingly, we identified VDAC1 (voltage-dependent anion channel 1) as a target for Parkin-mediated Lys 27 poly-ubiquitylation and mitophagy. Moreover, pathogenic Parkin mutations interfere with distinct steps of mitochondrial translocation, ubiquitylation and/or final clearance through mitophagy. Thus, our data provide functional links between PINK1, Parkin and the selective autophagy of mitochondria, which is implicated in the pathogenesis of Parkinson's disease.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy/physiology , Mitochondria/metabolism , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Voltage-Dependent Anion Channel 1/metabolism , Adaptor Proteins, Signal Transducing/genetics , Autophagy/genetics , Biological Transport/genetics , Biological Transport/physiology , Cell Line, Tumor , Fluorescent Antibody Technique , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Mutation , Parkinson Disease/genetics , Parkinson Disease/metabolism , Protein Kinases/genetics , RNA Interference , Sequestosome-1 Protein , Ubiquitin-Protein Ligases/genetics , Voltage-Dependent Anion Channel 1/genetics
12.
Diabetes ; 59(3): 670-8, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20009088

ABSTRACT

OBJECTIVE: The ubiquitin-proteasome system is the main degradation machinery for intracellularly altered proteins. Hyperglycemia has been shown to increase intracellular levels of the reactive dicarbonyl methylglyoxal (MGO) in cells damaged by diabetes, resulting in modification of proteins and alterations of their function. In this study, the influence of MGO-derived advanced glycation end product (AGE) formation on the activity of the proteasome was investigated in vitro and in vivo. RESEARCH DESIGN AND METHODS: MGO-derived AGE modification of proteasome subunits was analyzed by mass spectrometry, immunoprecipitation, and Western blots. Proteasome activity was analyzed using proteasome-specific fluorogenic substrates. Experimental models included bovine retinal endothelial cells, diabetic Ins2(Akita) mice, glyoxalase 1 (GLO1) knockdown mice, and streptozotocin (STZ)-injected diabetic mice. RESULTS: In vitro incubation with MGO caused adduct formation on several 20S proteasomal subunit proteins. In cultured endothelial cells, the expression level of the catalytic 20S proteasome subunit was not altered but proteasomal chymotrypsin-like activity was significantly reduced. In contrast, levels of regulatory 19S proteasomal proteins were decreased. In diabetic Ins2(Akita), STZ diabetic, and nondiabetic and diabetic G101 knockdown mice, chymotrypsin-like activity was also reduced and MGO modification of the 20S-beta2 subunit was increased. CONCLUSIONS: Hyperglycemia-induced formation of MGO covalently modifies the 20S proteasome, decreasing its activity in the diabetic kidney and reducing the polyubiquitin receptor 19S-S5a. The results indicate a new link between hyperglycemia and impairment of cell functions.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Endothelial Cells/metabolism , Hyperglycemia/metabolism , Proteasome Endopeptidase Complex/metabolism , Pyruvaldehyde/metabolism , Albumins/metabolism , Animals , Cattle , Cell Line , Chymotrypsin/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Female , Glucose/toxicity , Glycation End Products, Advanced/metabolism , Glycosylation , Humans , In Vitro Techniques , Insulin/genetics , Insulin/metabolism , Lactoylglutathione Lyase/genetics , Lactoylglutathione Lyase/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Polyubiquitin/metabolism
13.
Nucleic Acids Res ; 37(13): 4453-63, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19474347

ABSTRACT

DNA mismatch repair (MMR) and very-short patch (VSP) repair are two pathways involved in the repair of T:G mismatches. To learn about competition and cooperation between these two repair pathways, we analyzed the physical and functional interaction between MutL and Vsr using biophysical and biochemical methods. Analytical ultracentrifugation reveals a nucleotide-dependent interaction between Vsr and the N-terminal domain of MutL. Using chemical crosslinking, we mapped the interaction site of MutL for Vsr to a region between the N-terminal domains similar to that described before for the interaction between MutL and the strand discrimination endonuclease MutH of the MMR system. Competition between MutH and Vsr for binding to MutL resulted in inhibition of the mismatch-provoked MutS- and MutL-dependent activation of MutH, which explains the mutagenic effect of Vsr overexpression. Cooperation between MMR and VSP repair was demonstrated by the stimulation of the Vsr endonuclease in a MutS-, MutL- and ATP-hydrolysis-dependent manner, in agreement with the enhancement of VSP repair by MutS and MutL in vivo. These data suggest a mobile MutS-MutL complex in MMR signalling, that leaves the DNA mismatch prior to, or at the time of, activation of downstream effector molecules such as Vsr or MutH.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA Mismatch Repair , Endodeoxyribonucleases/metabolism , Escherichia coli Proteins/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/radiation effects , Cross-Linking Reagents , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases/chemistry , Endodeoxyribonucleases/radiation effects , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/radiation effects , MutL Proteins , MutS DNA Mismatch-Binding Protein/metabolism , Photochemical Processes , Protein Structure, Tertiary , Ultracentrifugation
SELECTION OF CITATIONS
SEARCH DETAIL