Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Michael Graf is active.

Publication


Featured researches published by Michael Graf.


Nature Structural & Molecular Biology | 2015

The proline-rich antimicrobial peptide Onc112 inhibits translation by blocking and destabilizing the initiation complex

A. Carolin Seefeldt; Fabian Nguyen; Stéphanie Antunes; Natacha Pérébaskine; Michael Graf; Stefan Arenz; K Kishore Inampudi; Céline Douat; Gilles Guichard; Daniel N. Wilson; C. Axel Innis

The increasing prevalence of multidrug-resistant pathogenic bacteria is making current antibiotics obsolete. Proline-rich antimicrobial peptides (PrAMPs) display potent activity against Gram-negative bacteria and thus represent an avenue for antibiotic development. PrAMPs from the oncocin family interact with the ribosome to inhibit translation, but their mode of action has remained unclear. Here we have determined a structure of the Onc112 peptide in complex with the Thermus thermophilus 70S ribosome at a resolution of 3.1 Å by X-ray crystallography. The Onc112 peptide binds within the ribosomal exit tunnel and extends toward the peptidyl transferase center, where it overlaps with the binding site for an aminoacyl-tRNA. We show biochemically that the binding of Onc112 blocks and destabilizes the initiation complex, thus preventing entry into the elongation phase. Our findings provide a basis for the future development of this class of potent antimicrobial agents.


Nature Communications | 2016

A combined cryo-EM and molecular dynamics approach reveals the mechanism of ErmBL-mediated translation arrest

Stefan Arenz; Lars V. Bock; Michael Graf; C. Axel Innis; Roland Beckmann; Helmut Grubmüller; Andrea C. Vaiana; Daniel N. Wilson

Nascent polypeptides can induce ribosome stalling, regulating downstream genes. Stalling of ErmBL peptide translation in the presence of the macrolide antibiotic erythromycin leads to resistance in Streptococcus sanguis. To reveal this stalling mechanism we obtained 3.6-Å-resolution cryo-EM structures of ErmBL-stalled ribosomes with erythromycin. The nascent peptide adopts an unusual conformation with the C-terminal Asp10 side chain in a previously unseen rotated position. Together with molecular dynamics simulations, the structures indicate that peptide-bond formation is inhibited by displacement of the peptidyl-tRNA A76 ribose from its canonical position, and by non-productive interactions of the A-tRNA Lys11 side chain with the A-site crevice. These two effects combine to perturb peptide-bond formation by increasing the distance between the attacking Lys11 amine and the Asp10 carbonyl carbon. The interplay between drug, peptide and ribosome uncovered here also provides insight into the fundamental mechanism of peptide-bond formation.


Nucleic Acids Research | 2016

Structure of the mammalian antimicrobial peptide Bac7(1-16) bound within the exit tunnel of a bacterial ribosome.

A. Carolin Seefeldt; Michael Graf; Natacha Pérébaskine; Fabian Nguyen; Stefan Arenz; Mario Mardirossian; Marco Scocchi; Daniel N. Wilson; C. Axel Innis

Proline-rich antimicrobial peptides (PrAMPs) produced as part of the innate immune response of animals, insects and plants represent a vast, untapped resource for the treatment of multidrug-resistant bacterial infections. PrAMPs such as oncocin or bactenecin-7 (Bac7) interact with the bacterial ribosome to inhibit translation, but their supposed specificity as inhibitors of bacterial rather than mammalian protein synthesis remains unclear, despite being key to developing drugs with low toxicity. Here, we present crystal structures of the Thermus thermophilus 70S ribosome in complex with the first 16 residues of mammalian Bac7, as well as the insect-derived PrAMPs metalnikowin I and pyrrhocoricin. The structures reveal that the mammalian Bac7 interacts with a similar region of the ribosome as insect-derived PrAMPs. Consistently, Bac7 and the oncocin derivative Onc112 compete effectively with antibiotics, such as erythromycin, which target the ribosomal exit tunnel. Moreover, we demonstrate that Bac7 allows initiation complex formation but prevents entry into the elongation phase of translation, and show that it inhibits translation on both mammalian and bacterial ribosomes, explaining why this peptide needs to be stored as an inactive pro-peptide. These findings highlight the need to consider the specificity of PrAMP derivatives for the bacterial ribosome in future drug development efforts.


Nature Structural & Molecular Biology | 2017

An antimicrobial peptide that inhibits translation by trapping release factors on the ribosome

Tanja Florin; Cristina Maracci; Michael Graf; Prajwal Karki; Dorota Klepacki; Otto Berninghausen; Roland Beckmann; Nora Vázquez-Laslop; Daniel N. Wilson; Marina V. Rodnina; Alexander S. Mankin

Many antibiotics stop bacterial growth by inhibiting different steps of protein synthesis. However, no specific inhibitors of translation termination are known. Proline-rich antimicrobial peptides, a component of the antibacterial defense system of multicellular organisms, interfere with bacterial growth by inhibiting translation. Here we show that Api137, a derivative of the insect-produced antimicrobial peptide apidaecin, arrests terminating ribosomes using a unique mechanism of action. Api137 binds to the Escherichia coli ribosome and traps release factor (RF) RF1 or RF2 subsequent to the release of the nascent polypeptide chain. A high-resolution cryo-EM structure of the ribosome complexed with RF1 and Api137 reveals the molecular interactions that lead to RF trapping. Api137-mediated depletion of the cellular pool of free release factors causes the majority of ribosomes to stall at stop codons before polypeptide release, thereby resulting in a global shutdown of translation termination.


Nucleic Acids Research | 2016

Cryo-EM structure of the spinach chloroplast ribosome reveals the location of plastid-specific ribosomal proteins and extensions

Michael Graf; Stefan Arenz; Paul Huter; Alexandra Dönhöfer; Jiří Nováček; Daniel N. Wilson

Abstract Ribosomes are the protein synthesizing machines of the cell. Recent advances in cryo-EM have led to the determination of structures from a variety of species, including bacterial 70S and eukaryotic 80S ribosomes as well as mitoribosomes from eukaryotic mitochondria, however, to date high resolution structures of plastid 70S ribosomes have been lacking. Here we present a cryo-EM structure of the spinach chloroplast 70S ribosome, with an average resolution of 5.4 Å for the small 30S subunit and 3.6 Å for the large 50S ribosomal subunit. The structure reveals the location of the plastid-specific ribosomal proteins (RPs) PSRP1, PSRP4, PSRP5 and PSRP6 as well as the numerous plastid-specific extensions of the RPs. We discover many features by which the plastid-specific extensions stabilize the ribosome via establishing additional interactions with surrounding ribosomal RNA and RPs. Moreover, we identify a large conglomerate of plastid-specific protein mass adjacent to the tunnel exit site that could facilitate interaction of the chloroplast ribosome with the thylakoid membrane and the protein-targeting machinery. Comparing the Escherichia coli 70S ribosome with that of the spinach chloroplast ribosome provides detailed insight into the co-evolution of RP and rRNA.


Molecular Cell | 2017

Structural Basis for Polyproline-Mediated Ribosome Stalling and Rescue by the Translation Elongation Factor EF-P

Paul Huter; Stefan Arenz; Lars V. Bock; Michael Graf; Jan Ole Frister; André Heuer; Lauri Peil; Agata L. Starosta; Ingo Wohlgemuth; Frank Peske; Jiří Nováček; Otto Berninghausen; Helmut Grubmüller; Tanel Tenson; Roland Beckmann; Marina V. Rodnina; Andrea C. Vaiana; Daniel N. Wilson

Ribosomes synthesizing proteins containing consecutive proline residues become stalled and require rescue via the action of uniquely modified translation elongation factors, EF-P in bacteria, or archaeal/eukaryotic a/eIF5A. To date, no structures exist of EF-P or eIF5A in complex with translating ribosomes stalled at polyproline stretches, and thus structural insight into how EF-P/eIF5A rescue these arrested ribosomes has been lacking. Here we present cryo-EM structures of ribosomes stalled on proline stretches, without and with modified EF-P. The structures suggest that the favored conformation of the polyproline-containing nascent chain is incompatible with the peptide exit tunnel of the ribosome and leads to destabilization of the peptidyl-tRNA. Binding of EF-P stabilizes the P-site tRNA, particularly via interactions between its modification and the CCA end, thereby enforcing an alternative conformation of the polyproline-containing nascent chain, which allows a favorable substrate geometry for peptide bond formation.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Structures of the orthosomycin antibiotics avilamycin and evernimicin in complex with the bacterial 70S ribosome

Stefan Arenz; Manuel F. Juette; Michael Graf; Fabian Nguyen; Paul Huter; Yury S. Polikanov; Scott C. Blanchard; Daniel N. Wilson

Significance The ribosome is the protein-synthesizing machine of the cell and is a major target for antibiotics. The increase in multidrug-resistant bacteria has limited the utility of our current arsenal of clinically used antibiotics, highlighting the need for further development of compounds that have distinct binding sites and do not display cross-resistance. Using cryo-electron microscopy, we have visualized the binding site of the orthosomycins evernimicin and avilamycin on the bacterial 70S ribosome. The binding site and mode of interaction of evernimicin and avilamycin are distinct from other ribosome-targeting antibiotics. Together with single-molecule studies, our structures reveal how the orthosomycin antibiotics inhibit protein synthesis by preventing accommodation of the aminoacyl-tRNA at the A site of the ribosome. The ribosome is one of the major targets for therapeutic antibiotics; however, the rise in multidrug resistance is a growing threat to the utility of our current arsenal. The orthosomycin antibiotics evernimicin (EVN) and avilamycin (AVI) target the ribosome and do not display cross-resistance with any other classes of antibiotics, suggesting that they bind to a unique site on the ribosome and may therefore represent an avenue for development of new antimicrobial agents. Here we present cryo-EM structures of EVN and AVI in complex with the Escherichia coli ribosome at 3.6- to 3.9-Å resolution. The structures reveal that EVN and AVI bind to a single site on the large subunit that is distinct from other known antibiotic binding sites on the ribosome. Both antibiotics adopt an extended conformation spanning the minor grooves of helices 89 and 91 of the 23S rRNA and interacting with arginine residues of ribosomal protein L16. This binding site overlaps with the elbow region of A-site bound tRNA. Consistent with this finding, single-molecule FRET (smFRET) experiments show that both antibiotics interfere with late steps in the accommodation process, wherein aminoacyl-tRNA enters the peptidyltransferase center of the large ribosomal subunit. These data provide a structural and mechanistic rationale for how these antibiotics inhibit the elongation phase of protein synthesis.


Proceedings of the FISITA 2012 World Automotive Congress | 2013

Torque Vectoring Control Design Based on Objective Driving Dynamic Parameters

Michael Graf; Markus Lienkamp

Due to the good controllability of electric motors the possibilities of driving behaviour control are increased. The amount of applicable yaw moment on the car compared to previous systems is increased; therefore it is essential to investigate the possibilities of influencing the driving experiences. Thus the subjective impression of the vehicle dynamics can be improved although the narrow roll-resistant optimized tyres on electric cars have less lateral potential than conventional ones. The objective of this study is to find the key driving dynamic parameters to evaluate the behaviour already in the simulation and to adjust the torque vectoring control unit to improve the drivability. The vehicle dynamics evaluation of the small electric car is based on characteristic values from literature research and handling tests. Further dynamic simulations and test drives with different torque vectoring target specifications generate a range for each parameter, where the driving behaviour for the driver is still on a high level. A correlation between the objective parameters gives a detailed overview of their specific importance in different driving manoeuvres. Depending on the uncontrolled vehicle behaviour a recommendation for the design criteria of the torque vectoring control unit is given. Thus in the time intensive driving tests only the control target (the yaw rate) has to be validated and adjusted.


Archaea | 2016

Deciphering the Translation Initiation Factor 5A Modification Pathway in Halophilic Archaea

Laurence Prunetti; Michael Graf; Ian K. Blaby; Lauri Peil; Andrea M. Makkay; Agata L. Starosta; R. Thane Papke; Tairo Oshima; Daniel N. Wilson; Valérie de Crécy-Lagard

Translation initiation factor 5A (IF5A) is essential and highly conserved in Eukarya (eIF5A) and Archaea (aIF5A). The activity of IF5A requires hypusine, a posttranslational modification synthesized in Eukarya from the polyamine precursor spermidine. Intracellular polyamine analyses revealed that agmatine and cadaverine were the main polyamines produced in Haloferax volcanii in minimal medium, raising the question of how hypusine is synthesized in this halophilic Archaea. Metabolic reconstruction led to a tentative picture of polyamine metabolism and aIF5A modification in Hfx. volcanii that was experimentally tested. Analysis of aIF5A from Hfx. volcanii by LC-MS/MS revealed it was exclusively deoxyhypusinylated. Genetic studies confirmed the role of the predicted arginine decarboxylase gene (HVO_1958) in agmatine synthesis. The agmatinase-like gene (HVO_2299) was found to be essential, consistent with a role in aIF5A modification predicted by physical clustering evidence. Recombinant deoxyhypusine synthase (DHS) from S. cerevisiae was shown to transfer 4-aminobutyl moiety from spermidine to aIF5A from Hfx. volcanii in vitro. However, at least under conditions tested, this transfer was not observed with the Hfx. volcanii DHS. Furthermore, the growth of Hfx. volcanii was not inhibited by the classical DHS inhibitor GC7. We propose a model of deoxyhypusine synthesis in Hfx. volcanii that differs from the canonical eukaryotic pathway, paving the way for further studies.


Proceedings of the National Academy of Sciences of the United States of America | 2018

Structural basis for antibiotic resistance mediated by the Bacillus subtilis ABCF ATPase VmlR

Caillan Crowe-McAuliffe; Michael Graf; Paul Huter; Hiraku Takada; Maha Abdelshahid; Jiří Nováček; Victoriia Murina; Gemma C. Atkinson; Vasili Hauryliuk; Daniel N. Wilson

Significance The recent increase in multidrug-resistant pathogenic bacteria is limiting the utility of our current arsenal of clinically important antibiotics. The development of improved antibiotics would therefore benefit from a better understanding of the current resistance mechanisms employed by bacteria. Many Gram-positive bacteria, including pathogenic Staphylococcus aureus and Enterococcus faecalis, utilize ribosome protection proteins to confer resistance to medically relevant antibiotics, such as streptogramins A, lincosamides, and pleuromutilins. We have employed cryo-electron microscopy to reveal the structural basis for how the Bacillus subtilis VmlR protein binds to the ribosome to confer resistance to the streptogramin A antibiotic virginiamycin M, the lincosamide lincomycin, and the pleuromutilin tiamulin. Many Gram-positive pathogenic bacteria employ ribosomal protection proteins (RPPs) to confer resistance to clinically important antibiotics. In Bacillus subtilis, the RPP VmlR confers resistance to lincomycin (Lnc) and the streptogramin A (SA) antibiotic virginiamycin M (VgM). VmlR is an ATP-binding cassette (ABC) protein of the F type, which, like other antibiotic resistance (ARE) ABCF proteins, is thought to bind to antibiotic-stalled ribosomes and promote dissociation of the drug from its binding site. To investigate the molecular mechanism by which VmlR confers antibiotic resistance, we have determined a cryo-electron microscopy (cryo-EM) structure of an ATPase-deficient B. subtilis VmlR-EQ2 mutant in complex with a B. subtilis ErmDL-stalled ribosomal complex (SRC). The structure reveals that VmlR binds within the E site of the ribosome, with the antibiotic resistance domain (ARD) reaching into the peptidyltransferase center (PTC) of the ribosome and a C-terminal extension (CTE) making contact with the small subunit (SSU). To access the PTC, VmlR induces a conformational change in the P-site tRNA, shifting the acceptor arm out of the PTC and relocating the CCA end of the P-site tRNA toward the A site. Together with microbiological analyses, our study indicates that VmlR allosterically dissociates the drug from its ribosomal binding site and exhibits specificity to dislodge VgM, Lnc, and the pleuromutilin tiamulin (Tia), but not chloramphenicol (Cam), linezolid (Lnz), nor the macrolide erythromycin (Ery).

Collaboration


Dive into the Michael Graf's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Roland Beckmann

Center for Integrated Protein Science Munich

View shared research outputs
Top Co-Authors

Avatar

Jiří Nováček

Central European Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alexander S. Mankin

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Dorota Klepacki

University of Illinois at Chicago

View shared research outputs
Researchain Logo
Decentralizing Knowledge