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










Publication year range
1.
PLoS One ; 18(12): e0290024, 2023.
Article in English | MEDLINE | ID: mdl-38157339

ABSTRACT

Epidermal cells of dark-grown plant seedlings reorient their cortical microtubule arrays in response to blue light from a net lateral orientation to a net longitudinal orientation with respect to the long axis of cells. The molecular mechanism underlying this microtubule array reorientation involves katanin, a microtubule severing enzyme, and a plant-specific microtubule associated protein called SPIRAL2. Katanin preferentially severs longitudinal microtubules, generating seeds that amplify the longitudinal array. Upon severing, SPIRAL2 binds nascent microtubule minus ends and limits their dynamics, thereby stabilizing the longitudinal array while the lateral array undergoes net depolymerization. To date, no experimental structural information is available for SPIRAL2 to help inform its mechanism. To gain insight into SPIRAL2 structure and function, we determined a 1.8 Å resolution crystal structure of the Arabidopsis thaliana SPIRAL2 C-terminal domain. The domain is composed of seven core α-helices, arranged in an α-solenoid. Amino-acid sequence conservation maps primarily to one face of the domain involving helices α1, α3, α5, and an extended loop, the α6-α7 loop. The domain fold is similar to, yet structurally distinct from the C-terminal domain of Ge-1 (an mRNA decapping complex factor involved in P-body localization) and, surprisingly, the C-terminal domain of the katanin p80 regulatory subunit. The katanin p80 C-terminal domain heterodimerizes with the MIT domain of the katanin p60 catalytic subunit, and in metazoans, binds the microtubule minus-end factors CAMSAP3 and ASPM. Structural analysis predicts that SPIRAL2 does not engage katanin p60 in a mode homologous to katanin p80. The SPIRAL2 structure highlights an interesting evolutionary convergence of domain architecture and microtubule minus-end localization between SPIRAL2 and katanin complexes, and establishes a foundation upon which structure-function analysis can be conducted to elucidate the role of this domain in the regulation of plant microtubule arrays.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Amino Acid Sequence , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Katanin/chemistry , Microtubules/metabolism , Plants/metabolism
2.
Biomolecules ; 13(4)2023 03 30.
Article in English | MEDLINE | ID: mdl-37189368

ABSTRACT

In cells, microtubule location, length, and dynamics are regulated by a host of microtubule-associated proteins and enzymes that read where to bind and act based on the microtubule "tubulin code," which is predominantly encoded in the tubulin carboxy-terminal tail (CTT). Katanin is a highly conserved AAA ATPase enzyme that binds to the tubulin CTTs to remove dimers and sever microtubules. We have previously demonstrated that short CTT peptides are able to inhibit katanin severing. Here, we examine the effects of CTT sequences on this inhibition activity. Specifically, we examine CTT sequences found in nature, alpha1A (TUBA1A), detyrosinated alpha1A, Δ2 alpha1A, beta5 (TUBB/TUBB5), beta2a (TUBB2A), beta3 (TUBB3), and beta4b (TUBB4b). We find that these natural CTTs have distinct abilities to inhibit, most noticeably beta3 CTT cannot inhibit katanin. Two non-native CTT tail constructs are also unable to inhibit, despite having 94% sequence identity with alpha1 or beta5 sequences. Surprisingly, we demonstrate that poly-E and poly-D peptides are capable of inhibiting katanin significantly. An analysis of the hydrophobicity of the CTT constructs indicates that more hydrophobic polypeptides are less inhibitory than more polar polypeptides. These experiments not only demonstrate inhibition, but also likely interaction and targeting of katanin to these various CTTs when they are part of a polymerized microtubule filament.


Subject(s)
Microtubules , Tubulin , Tubulin/metabolism , Katanin/analysis , Katanin/chemistry , Katanin/metabolism , Microtubules/metabolism , Microtubule-Associated Proteins/metabolism
3.
Acta Crystallogr F Struct Biol Commun ; 79(Pt 1): 17-22, 2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36598352

ABSTRACT

Plant cells form microtubule arrays, called `cortical microtubules', beneath the plasma membrane which are critical for cell-wall organization and directional cell growth. Cortical microtubules are nucleated independently of centrosomes. Spiral2 is a land-plant-specific microtubule minus-end-targeting protein that stabilizes the minus ends by inhibiting depolymerization of the filament. Spiral2 possesses an N-terminal microtubule-binding domain and a conserved C-terminal domain whose function is unknown. In this study, the crystal structure of the conserved C-terminal domain of Spiral2 was determined using the single-wavelength anomalous dispersion method. Refinement of the model to a resolution of 2.2 Šrevealed a helix-turn-helix fold with seven α-helices. The protein crystallized as a dimer, but SEC-MALS analysis showed the protein to be monomeric. A structural homology search revealed that the protein has similarity to the C-terminal domain of the katanin regulatory subunit p80. The structure presented here suggests that the C-terminal domain of Spiral2 represents a new class of microtubule dynamics modulator across the kingdom.


Subject(s)
Microtubule-Associated Proteins , Microtubules , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Crystallography, X-Ray , Microtubules/metabolism , Katanin/chemistry , Katanin/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism
4.
J Phys Chem B ; 126(50): 10569-10586, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36475672

ABSTRACT

Severing proteins are nanomachines from the AAA+ (ATPases associated with various cellular activities) superfamily whose function is to remodel the largest cellular filaments, microtubules. The standard AAA+ machines adopt hexameric ring structures for functional reasons, while being primarily monomeric in the absence of the nucleotide. Both major severing proteins, katanin and spastin, are believed to follow this trend. However, studies proposed that they populate lower-order oligomers in the presence of cofactors, which are functionally relevant. Our simulations show that the preferred oligomeric assembly is dependent on the binding partners and on the type of severing protein. Essential dynamics analysis predicts that the stability of an oligomer is dependent on the strength of the interface between the helical bundle domain (HBD) of a monomer and the convex face of the nucleotide binding domain (NBD) of a neighboring monomer. Hot spots analysis found that the region consisting of the HBD tip and the C-terminal (CT) helix is the only common element between the allosteric networks responding to nucleotide, substrate, and intermonomer binding. Clustering analysis indicates the existence of multiple pathways for the transition between the secondary structure of the HBD tip in monomers and the structure(s) it adopts in oligomers.


Subject(s)
Adenosine Triphosphatases , Microtubules , Katanin/chemistry , Katanin/metabolism , Spastin/metabolism , Adenosine Triphosphatases/chemistry , Nucleotides/metabolism
5.
Dev Cell ; 52(1): 118-131.e6, 2020 01 06.
Article in English | MEDLINE | ID: mdl-31735665

ABSTRACT

The AAA ATPase katanin severs microtubules. It is critical in cell division, centriole biogenesis, and neuronal morphogenesis. Its mutation causes microcephaly. The microtubule templates katanin hexamerization and activates its ATPase. The structural basis for these activities and how they lead to severing is unknown. Here, we show that ß-tubulin tails are necessary and sufficient for severing. Cryoelectron microscopy (cryo-EM) structures reveal the essential tubulin tail glutamates gripped by a double spiral of electropositive loops lining the katanin central pore. Each spiral couples allosterically to the ATPase and binds alternating, successive substrate residues, with consecutive residues coordinated by adjacent protomers. This tightly couples tail binding, hexamerization, and ATPase activation. Hexamer structures in different states suggest an ATPase-driven, ratchet-like translocation of the tubulin tail through the pore. A disordered region outside the AAA core anchors katanin to the microtubule while the AAA motor exerts the forces that extract tubulin dimers and sever the microtubule.


Subject(s)
Caenorhabditis elegans/metabolism , Katanin/metabolism , Microtubules/metabolism , Tubulin/metabolism , Adenosine Triphosphate/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Humans , Katanin/chemistry , Katanin/genetics , Models, Molecular , Protein Conformation , Tubulin/chemistry , Tubulin/genetics
6.
Structure ; 27(9): 1375-1383.e3, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31353241

ABSTRACT

Katanin is a microtubule-severing enzyme that is crucial for many cellular processes. Katanin consists of two subunits, p60 and p80, that form a stable complex. The interaction between subunits is mediated by the p60 N-terminal microtubule-interacting and -trafficking domain (p60-MIT) and the p80 C-terminal domain (p80-CTD). Here, we performed a biophysical characterization of the mouse p60-MIT:p80-CTD heterodimer and show that this complex can assemble into heterotetramers. We identified two mutations that enhance heterotetramer formation and determined the X-ray crystal structure of this mutant complex. The structure revealed a domain-swapped heterotetramer consisting of two p60-MIT:p80-CTD heterodimers. Structure-based sequence alignments suggest that heterotetramerization of katanin might be a common feature of various species. Furthermore, we show that enhanced heterotetramerization of katanin impairs its microtubule end-binding properties and increases the enzyme's microtubule lattice binding and severing activities. Therefore, our findings suggest the existence of different katanin oligomers that possess distinct functional properties.


Subject(s)
Katanin/chemistry , Mutation , Animals , Binding Sites , Crystallography, X-Ray , Katanin/genetics , Katanin/metabolism , Mice , Microtubules/metabolism , Models, Molecular , Protein Conformation , Protein Domains , Protein Multimerization , Sequence Alignment
7.
PLoS One ; 14(2): e0212518, 2019.
Article in English | MEDLINE | ID: mdl-30789974

ABSTRACT

Microtubule severing, which is highly critical for the survival of both mitotic and post-mitotic cells, has to be precisely adjusted by regulating the expression levels of severing proteins, katanin and spastin. Even though severing mechanism is relatively well-studied, there are limited studies for the transcriptional regulation of microtubule severing proteins. In this study, we identified the main regulatory region of KATNA1 gene encoding katanin-p60 as 5' UTR, which has a key role for its expression, and showed Elk1 binding to KATNA1. Furthermore, we identified that Elk1 decreased katanin-p60 and spastin protein expressions, while mRNA levels were increased upon Elk1 overexpression. In addition, SUMOylation is a known post-translational modification regulating Elk1 activity. A previous study suggested that K230, K249, K254 amino acids in the R domain are the main SUMOylation sites; however, we identified that these amino acids are neither essential nor substantial for Elk1 SUMOylation. Also, we determined that KATNA1 methylation results in the reduction of Elk1 binding whereas SPG4 methylation does not. Together, our findings emphasizing the impacts of both transcriptional and post-transcriptional regulations of katanin-p60 and spastin suggest that Elk1 has a key role for differential expression patterns of microtubule severing proteins, thereby regulating cellular functions through alterations of microtubule organization.


Subject(s)
Katanin/metabolism , Spastin/metabolism , ets-Domain Protein Elk-1/metabolism , 5' Untranslated Regions , Amino Acid Sequence , Amino Acid Substitution , Binding Sites , Cell Line , DNA Methylation , Humans , Katanin/chemistry , Katanin/genetics , Microtubules/genetics , Microtubules/metabolism , Mutagenesis, Site-Directed , Protein Interaction Domains and Motifs , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spastin/chemistry , Spastin/genetics , Sumoylation , Transcription, Genetic , ets-Domain Protein Elk-1/chemistry , ets-Domain Protein Elk-1/genetics
8.
Cell Rep ; 26(5): 1357-1367.e5, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30699360

ABSTRACT

Katanin was the first microtubule (MT)-severing enzyme discovered, but how katanin executes MT severing remains poorly understood. Here, we report X-ray crystal structures of the apo and ATPγS-bound states of the catalytic AAA domain of human katanin p60 at 3.0 and 2.9 Å resolution, respectively. Comparison of the two structures reveals conformational changes induced by ATP binding and how such changes ensure hexamer stability. Moreover, we uncover structural details of pore loops (PLs) and show that Arg283, a residue unique to katanin among MT-severing enzymes, protrudes from PL1 and lines the entry of the catalytic pore. Functional studies suggest that PL1 and Arg283 play essential roles in the recognition and remodeling of the glutamylated, C-terminal tubulin tail and regulation of axon growth. In addition, domain-swapping experiments in katanin and spastin suggest that the non-homologous N-terminal region, which contains the MT-interacting and trafficking domain and a linker, confers specificity to the severing process.


Subject(s)
Glutamates/metabolism , Katanin/chemistry , Katanin/metabolism , Microtubules/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Arginine/metabolism , Axons/metabolism , HeLa Cells , Humans , Mice, Inbred ICR , Models, Molecular , Mutation/genetics , Protein Domains , Protein Multimerization , Sensory Receptor Cells/metabolism , Spastin/metabolism
9.
J Biol Chem ; 293(27): 10590-10605, 2018 07 06.
Article in English | MEDLINE | ID: mdl-29752405

ABSTRACT

The reorganization of microtubules in mitosis, meiosis, and development requires the microtubule-severing activity of katanin. Katanin is a heterodimer composed of an ATPase associated with diverse cellular activities (AAA) subunit and a regulatory subunit. Microtubule severing requires ATP hydrolysis by katanin's conserved AAA ATPase domains. Whereas other AAA ATPases form stable hexamers, we show that katanin forms only a monomer or dimers of heterodimers in solution. Katanin oligomers consistent with hexamers of heterodimers or heterododecamers were only observed for an ATP hydrolysis-deficient mutant in the presence of ATP. X-ray structures of katanin's AAA ATPase in monomeric nucleotide-free and pseudo-oligomeric ADP-bound states revealed conformational changes in the AAA subdomains that explained the structural basis for the instability of the katanin heterododecamer. We propose that the rapid dissociation of katanin AAA oligomers may lead to an autoinhibited state that prevents inappropriate microtubule severing or that cyclical disassembly into heterodimers may critically contribute to the microtubule-severing mechanism.


Subject(s)
Adenosine Triphosphatases/chemistry , Caenorhabditis elegans Proteins/chemistry , Katanin/chemistry , Meiosis , Adenosine Triphosphatases/metabolism , Animals , Caenorhabditis elegans/enzymology , Caenorhabditis elegans Proteins/metabolism , Crystallography, X-Ray , Humans , Katanin/metabolism , Microtubules , Protein Conformation , Protein Multimerization , Spindle Apparatus
10.
Structure ; 26(3): 375-382.e4, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29395789

ABSTRACT

CAMSAP/Patronin family members regulate the organization and stability of microtubule minus ends in various systems ranging from mitotic spindles to differentiated epithelial cells and neurons. Mammalian CAMSAP2 and CAMSAP3 bind to growing microtubule minus ends, where they form stretches of stabilized microtubule lattice. The microtubule-severing ATPase katanin interacts with CAMSAPs and limits the length of CAMSAP-decorated microtubule stretches. Here, by using biochemical, biophysical, and structural approaches, we reveal that a short helical motif conserved in CAMSAP2 and CAMSAP3 binds to the heterodimer formed by the N- and C-terminal domains of katanin subunits p60 and p80, respectively. The identified CAMSAP-katanin binding mode is supported by mutational analysis and genome-editing experiments. It is strikingly similar to the one seen in the ASPM-katanin complex, which is responsible for microtubule minus-end regulation in mitotic spindles. Our work provides a general molecular mechanism for the cooperation of katanin with major microtubule minus-end regulators.


Subject(s)
Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , Katanin/chemistry , Katanin/metabolism , Animals , Binding Sites , Cytoskeletal Proteins/genetics , Gene Editing , Humans , Katanin/genetics , Mice , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Protein Binding , Protein Conformation , Protein Domains
11.
Sci Rep ; 7(1): 14893, 2017 11 02.
Article in English | MEDLINE | ID: mdl-29097679

ABSTRACT

Interactions between microtubule (MT) interacting and trafficking (MIT) domains and their binding proteins are important for the accurate progression of many cellular processes that require the AAA+ ATPase machinery. Therefore, knowledge on the structural basis of MIT domain interactions is crucial for understanding the molecular mechanisms underlying AAA+ ATPase function. Katanin is a MT-severing AAA+ ATPase that consists of p60 and p80 subunits. Although, the hexameric p60 subunit is active alone, its association with the p80 subunit greatly enhances both the MT-binding and -severing activities of katanin. However, the molecular mechanism of how the p80 subunit contributes to katanin function is currently unknown. Here, we structurally and functionally characterized the interaction between the two katanin subunits that is mediated by the p60-MIT domain and the p80 C-terminal domain (p80-CTD). We show that p60-MIT and p80-CTD form a tight heterodimeric complex, whose high-resolution structure we determined by X-ray crystallography. Based on the crystal structure, we identified two conserved charged residues that are important for p60-MIT:p80-CTD complex formation and katanin function. Moreover, p60-MIT was compared with other MIT domain structures and similarities are discussed.


Subject(s)
Katanin/metabolism , Animals , Crystallography, X-Ray , Katanin/chemistry , Mice , Models, Molecular , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Protein Interaction Maps , Protein Multimerization
12.
EMBO J ; 36(23): 3435-3447, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28978669

ABSTRACT

The microtubule (MT)-severing enzyme katanin triggers dynamic reorientation of cortical MT arrays that play crucial functions during plant cell morphogenesis, such as cell elongation, cell wall biosynthesis, and hormonal signaling. MT severing specifically occurs at crossover or branching nucleation sites in living Arabidopsis cells. This differs from the random severing observed along the entire length of single MTs in vitro and strongly suggests that a precise control mechanism must exist in vivo However, how katanin senses and cleaves at MT crossover and branching nucleation sites in vivo has remained unknown. Here, we show that the katanin p80 subunit KTN80 confers precision to MT severing by specific targeting of the katanin p60 subunit KTN1 to MT cleavage sites and that KTN1 is required for oligomerization of functional KTN80-KTN1 complexes that catalyze severing. Moreover, our findings suggest that the katanin complex in Arabidopsis is composed of a hexamer of KTN1-KTN80 heterodimers that sense MT geometry to confer precise MT severing. Our findings shed light on the precise control mechanism of MT severing in plant cells, which may be relevant for other eukaryotes.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Katanin/metabolism , Arabidopsis/genetics , Arabidopsis/growth & development , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Genes, Plant , Katanin/chemistry , Katanin/genetics , Microtubules/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Mutation , Plants, Genetically Modified , Protein Structure, Quaternary , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism
13.
Postepy Biochem ; 62(1): 46-51, 2016.
Article in Polish | MEDLINE | ID: mdl-28132444

ABSTRACT

Microtubule severing proteins, katanin, spastin and fidgetin cause local destabilization of the microtubules structure. This ATP-dependent activity leads to the shortening or disassembly of the existing microtubules. The generated short microtubule fragments may serve as templates to polymerize new microtubules and in consequence, the activity of the microtubule severing proteins leads to the reorganization of the microtubular cytoskeleton. This review summarizes current knowledge concerning structural organization of the microtubule severing proteins, the molecular mechanism of their action, factors that regulate the level of the katanin and spastin within the cells and their microtubule severing activity.


Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , Katanin/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Spastin/metabolism , ATPases Associated with Diverse Cellular Activities/chemistry , Adenosine Triphosphate/metabolism , Animals , Humans , Katanin/chemistry , Microtubule-Associated Proteins/chemistry , Protein Conformation , Spastin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL