Structural Mechanisms for VMAT2 inhibition by tetrabenazine

  1. Department of Structural Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
  2. Laufer Center for Physical and Quantitative Biology, and Department of Biochemistry and Cell Biology, School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA

Editors

  • Reviewing Editor
    Shimon Schuldiner
    The Hebrew University of Jerusalem, Jerusalem, Israel
  • Senior Editor
    Merritt Maduke
    Stanford University, Stanford, United States of America

Reviewer #1 (Public Review):

Summary: This study presents fundamental new insights into vesicular monoamine transport and the binding pose of the clinical drug tetrabenazine (TBZ) to the mammalian VMAT2 transporter. Specifically, this study reports the first structure for the mammalian VMAT (SLC18) family of vesicular monoamine transporters. It provides insights into the mechanism by which this inhibitor traps VMAT2 into a 'dead-end' conformation. The structure also provides some evidence for a novel gating mechanism within VMAT2, which may have wider implications for understanding the mechanism of transport in the wider SLC18 family.

Strengths: The structure is high quality, and the method used to determine the structure via fusing mVenus and the anti-GFP nanobody to the amino and carboxyl termini is novel. The binding and transport data are convincing, although limited. The binding position of TBZ is of high value, given its role in treating Huntington's chorea and for being a 'dead-end' inhibitor for VMAT2.

Weaknesses: The lack of additional mutational data and/or analyses on the impact of pH on ligand binding reduces the insights from these experiments. This reduces the strength of the conclusions that can be drawn about the mechanism of binding and transport or the novelty of the gating mechanism discussed above.

Reviewer #2 (Public Review):

Overview:

As a report of the first structure of VMAT2, indeed the first structure of any vesicular monoamine transporter, this manuscript represents an important milestone in the field of neurotransmitter transport. VMAT2 belongs to a large family (the major facilitator superfamily, MFS) containing transporters from all living species. There is a wealth of information relating to the way that MFS transporters bind substrates, undergo conformational changes to transport them across the membrane, and couple these events to the transmembrane movement of ions. VMAT2 couples the movement of protons out of synaptic vesicles to the vesicular uptake of biogenic amines (serotonin, dopamine, and norepinephrine) from the cytoplasm. The new structure presented in this manuscript can be expected to contribute to an understanding of this proton/amine antiport process.

The structure contains a molecule of the inhibitor TBZ bound in a central cavity, with no access to either luminal or cytoplasmic compartments. The authors carefully analyze which residues interact with bound TBZ and measure TBZ binding to VMAT2 mutated at some of those residues. These measurements allow well-reasoned conclusions about the differences in inhibitor selectivity between VMAT1 and VMAT2 and differences in affinity between TBZ derivatives.

The structure also reveals polar networks within the protein and hydrophobic residues in positions that may allow them to open and close pathways between the central binding site and the cytoplasm or the vesicle lumen. The authors propose the involvement of these networks and hydrophobic residues in the coupling of transport to proton translocation and conformational changes. However, these proposals are quite speculative in the absence of supporting structures and experimentation that would test specific mechanistic details.

Critique:

Although the structure presented in this MS is clearly important, I feel that the authors have overstated several of the conclusions that can be drawn from it. I don't agree that the structure clearly indicates why TBZ is a non-competitive inhibitor; the proposal that specific hydrophobic residues function as gates will depend on lumen- and cytoplasm-facing structures for verification; the polar networks could have any number of functions - indeed it would be surprising if they were all involved in proton transport. Several of these issues could be resolved by a clearer illustration of the data, but I believe that a more rigorous description of the conclusions and where they fall between firm findings and speculation would help the reader put the results in perspective.

Non-competitive inhibition occurs when the action of an inhibitor can't be overcome by increasing substrate concentration. The structure shows TBZ sequestered in the central cavity with no access to either cytoplasm or lumen. The explanation of competitive vs non-competitive inhibition depends entirely on how TBZ got there. If it is bound from the cytoplasm, cytoplasmic substrate should have been able to compete with TBZ and overcome the inhibition. If it is bound from the lumen, or from within the bilayer, cytoplasmic substrate would not be able to compete, and inhibition would be non-competitive. The structure does not tell us how TBZ got there, only that it was eventually occluded from both aqueous compartments and the bilayer.

The issue of how VMAT2 opens access to the central binding site from luminal and cytoplasmic sides is an important and interesting one, and comparison with other MFS structures in cytoplasmic-open or extracellular/luminal-open is a very reasonable approach. However, any conclusions for VMAT2 should be clearly indicated as speculative in the absence of comparable open structures of VMAT2. As a matter of presentation, I found the illustrations in ED Fig. 6 to be less helpful than they could have been. Specifically, illustrations that focus on the proposed gates, comparing that region of the new structure with the corresponding region of either VGLUT or GLUT4 would better help the reader to compare the position of the proposed gate residues with the corresponding region of the open structure. I realize that is the intended purpose of ED Fig. 6b and 6c, but currently, those show the entire protein, and a focus on the gate regions might make the proposed gate movements clearer. I also appreciate the difference between the Alphafold prediction and the new structure, but I'm not convinced that ED Fig. 6a adds anything helpful.

The polar networks described in the manuscript provide interesting possibilities for interactions with substrates and protons whose binding to VMAT2 must control conformational change. Aside from the description of these networks, there is little evidence presented to assess the role of these networks in transport. Are the networks conserved in other closely related transporters? How could the interaction of the networks with substrate or protons affect conformational change? Of course, any potential role proposed for the networks would be highly speculative at this point, and any discussion of their role should point out their speculative nature and the need for experimental verification. Some speculation, however, can be useful for focusing the field's attention on future directions. However, statements in the abstract (three distinct polar networks... play a role in proton transduction.) and the discussion (...are likely also involved in mediating proton transduction.) should be clearly presented as speculation until they are validated experimentally.

The strongest aspect of this work (aside from the structure itself) is the analysis of TBZ binding. There is a problematic aspect to this analysis. The discussion on how TBZ stabilizes the occluded conformation of VMAT2 is premature without structures of apo-VMAT2 and possibly structures with other ligands bound. We don't really know at this point whether VMAT2 might be in the same occluded conformation in the absence of TBZ. Any statements regarding the effect of interactions between VMAT2 and TBZ depend on demonstrating that TBZ has a conformational effect. The same applies to the discussion of the role of W318 on conformation and to the loops proposed to "occlude the luminal side of the transporter" (line 131).

The description of VMAT2 mechanism makes many assumptions that are based on studies with other MFS transporters. Rather than stating these assumptions as fact (VMAT2 functions by alternating access...), it would be preferable to explain why a reader should believe these assumptions. In general, this discussion presents conclusions as established facts rather than proposals that need to be tested experimentally.

The MD simulations are not described well enough for a general reader. What is the significance of the different runs? ED Fig. 4d is not high enough resolution to see the details.

Reviewer #3 (Public Review):

Summary:

The vesicular monoamine transporter is a key component in neuronal signaling and is implicated in diseases such as Parkinson's. Understanding of monoamine processing and our ability to target that process therapeutically has been to date provided by structural modeling and extensive biochemical studies. However, structural data is required to establish these findings more firmly.

Strengths:

Dalton et al resolved a structure of VMAT2 in the presence of an important inhibitor, tetrabenazine, with the protein in detergent micelles, using cryo-EM and with the aid of domains fused to its N- and C-terminal ends. The resolution of the maps allows clear assignment of the amino acids in the core of the protein. The structure is in good agreement with a wealth of experimental and structural prediction data and provides important insights into the binding site for tetrabenazine and selectivity relative to analogous compounds.

Weaknesses:

The authors follow up their structures with molecular dynamics simulations. The simulations resulted in repositioning of the ligand, which does not seem to be well founded, and raises questions about the methodological choices made for the simulations.

Author Response

Reviewer #1 (Public Review):

Summary: This study presents fundamental new insights into vesicular monoamine transport and the binding pose of the clinical drug tetrabenazine (TBZ) to the mammalian VMAT2 transporter. Specifically, this study reports the first structure for the mammalian VMAT (SLC18) family of vesicular monoamine transporters. It provides insights into the mechanism by which this inhibitor traps VMAT2 into a 'dead-end' conformation. The structure also provides some evidence for a novel gating mechanism within VMAT2, which may have wider implications for understanding the mechanism of transport in the wider SLC18 family.

Strengths: The structure is high quality, and the method used to determine the structure via fusing mVenus and the anti-GFP nanobody to the amino and carboxyl termini is novel. The binding and transport data are convincing, although limited. The binding position of TBZ is of high value, given its role in treating Huntington's chorea and for being a 'dead-end' inhibitor for VMAT2.

Weaknesses: The lack of additional mutational data and/or analyses on the impact of pH on ligand binding reduces the insights from these experiments. This reduces the strength of the conclusions that can be drawn about the mechanism of binding and transport or the novelty of the gating mechanism discussed above.

We greatly appreciate this summary and thank reviewer #1 for their comments and suggested experiments which we believe will further strengthen this work. We agree with these comments and plan to include more mutagenesis data in a revised manuscript in order to address this point and expand further on the mechanistic details of transport.

Reviewer #2 (Public Review):

Overview:

As a report of the first structure of VMAT2, indeed the first structure of any vesicular monoamine transporter, this manuscript represents an important milestone in the field of neurotransmitter transport. VMAT2 belongs to a large family (the major facilitator superfamily, MFS) containing transporters from all living species. There is a wealth of information relating to the way that MFS transporters bind substrates, undergo conformational changes to transport them across the membrane, and couple these events to the transmembrane movement of ions. VMAT2 couples the movement of protons out of synaptic vesicles to the vesicular uptake of biogenic amines (serotonin, dopamine, and norepinephrine) from the cytoplasm. The new structure presented in this manuscript can be expected to contribute to an understanding of this proton/amine antiport process.

The structure contains a molecule of the inhibitor TBZ bound in a central cavity, with no access to either luminal or cytoplasmic compartments. The authors carefully analyze which residues interact with bound TBZ and measure TBZ binding to VMAT2 mutated at some of those residues. These measurements allow well-reasoned conclusions about the differences in inhibitor selectivity between VMAT1 and VMAT2 and differences in affinity between TBZ derivatives.

The structure also reveals polar networks within the protein and hydrophobic residues in positions that may allow them to open and close pathways between the central binding site and the cytoplasm or the vesicle lumen. The authors propose the involvement of these networks and hydrophobic residues in the coupling of transport to proton translocation and conformational changes. However, these proposals are quite speculative in the absence of supporting structures and experimentation that would test specific mechanistic details.

Thank you for these comments and summary describing this work. We agree that the involvement of polar networks has not been experimentally tested; these are proposed as a possible mechanism, but we have not made mechanistic conclusions on how protons are translocated and coupled to transport. We believe we have made it clear in the manuscript when describing the polar networks that the corresponding discussion is largely descriptive and speculative and will further stress that in a future revision. We would like to point out however, that many of the polar and charged residues which make up these networks have been studied and that there is a wealth of biochemical and functional experiments in the literature which implicate these residues in this process. Yet, we agree that establishing the precise mechanistic details will require additional structures and likely also extensive computational experiments. We have cited these papers that have characterized these polar residues extensively throughout the text (30-32,37,49,55).

We would like to submit that we have not proposed that the hydrophobic gates are involved in proton translocation. Gating residues, by definition, block access to the binding site (29,30,48); and since our structure is occluded, we directly observe the residues which participate in both gates. We have also performed extensive mutagenesis studies of many of these hydrophobic gating residues and our binding data are consistent with this conclusion. Transport experiments with mutations at these gates might be helpful toward gaining a deeper understanding of transport mechanism but given the current structural data it is conceivable that these residues play a role in gating neurotransmitter.

Critique:

Although the structure presented in this MS is clearly important, I feel that the authors have overstated several of the conclusions that can be drawn from it. I don't agree that the structure clearly indicates why TBZ is a non-competitive inhibitor; the proposal that specific hydrophobic residues function as gates will depend on lumen- and cytoplasm-facing structures for verification; the polar networks could have any number of functions - indeed it would be surprising if they were all involved in proton transport. Several of these issues could be resolved by a clearer illustration of the data, but I believe that a more rigorous description of the conclusions and where they fall between firm findings and speculation would help the reader put the results in perspective.

The central argument made by this reviewer that is repeated throughout this critique is that more structures of various states are needed to make mechanistic conclusions with respect to how TBZ binds and alternating access. While additional structures would certainly add mechanistic detail, they are not required to make these conclusions. In fact, as we point out throughout the text, these conclusions have already been made in various publications which we have cited and discussed. Decades of mutagenesis, binding, transport, inhibition, and accessibility measurements all support the conclusion that TBZ binds from the luminal side and that VMAT2 uses an alternating mechanism to transport neurotransmitter (30-32,35-37,55). Structures are neither required nor sufficient to make such claims and more structures of various apo states in different conformations would not provide any additional support to this question. If the predominant apo state was luminal open, cytoplasm open or occluded, this would not prove how TBZ enters VMAT2. Structural data alone does not provide these details; biochemical data does and structures are useful for understanding the details of how these mechanisms work. Thus, our structure provides the molecular framework for understanding the binding site, conformation, gating, and polar networks and we have interpreted our own biochemical data as well as the available biochemical data in the literature in the context of our structure.

The structure indicates why TBZ is a non-competitive inhibitor (35,36) because it is not possible for neurotransmitters to compete for binding to this state. Neurotransmitter initially binds to the cytosolic facing state where the intracellular gates are open, inhibition by binding to this state would result in a competitive mechanism. Since TBZ is non-competitive, it must bind through the luminal-open state where the luminal gate is open. Further conformational change produces the occluded conformation with both the luminal and intracellular gates closed which is what we observe in the structure. This finding is supported by numerous biochemical and functional experiments and by extensive analysis of mutants in the gates using binding assays, transport experiments and cysteine accessibility experiments. We have cited and discussed these key papers (30-32,35-37,55) throughout the text and our results support the conclusions drawn from these works.

Non-competitive inhibition occurs when the action of an inhibitor can't be overcome by increasing substrate concentration. The structure shows TBZ sequestered in the central cavity with no access to either cytoplasm or lumen. The explanation of competitive vs non-competitive inhibition depends entirely on how TBZ got there. If it is bound from the cytoplasm, cytoplasmic substrate should have been able to compete with TBZ and overcome the inhibition. If it is bound from the lumen, or from within the bilayer, cytoplasmic substrate would not be able to compete, and inhibition would be non-competitive. The structure does not tell us how TBZ got there, only that it was eventually occluded from both aqueous compartments and the bilayer.

TBZ is accepted to be a non-competitive inhibitor, based on decades of research, and not based solely on our structure (30-32,35,36). Our structure provides insight into the molecular mechanism by which non-competitive inhibition occurs. Previous studies have shown that TBZ enters through the luminal side of the transporter, resulting in non-competitive inhibition by binding to a conformation of the transporter which does not bind cytosolic neurotransmitter. We agree our structure does not prove how TBZ ‘got there’, but other studies have addressed this question (30-32, 35, 36) and have been discussed in detail.

The issue of how VMAT2 opens access to the central binding site from luminal and cytoplasmic sides is an important and interesting one, and comparison with other MFS structures in cytoplasmic-open or extracellular/luminal-open is a very reasonable approach. However, any conclusions for VMAT2 should be clearly indicated as speculative in the absence of comparable open structures of VMAT2. As a matter of presentation, I found the illustrations in ED Fig. 6 to be less helpful than they could have been. Specifically, illustrations that focus on the proposed gates, comparing that region of the new structure with the corresponding region of either VGLUT or GLUT4 would better help the reader to compare the position of the proposed gate residues with the corresponding region of the open structure. I realize that is the intended purpose of ED Fig. 6b and 6c, but currently, those show the entire protein, and a focus on the gate regions might make the proposed gate movements clearer. I also appreciate the difference between the Alphafold prediction and the new structure, but I'm not convinced that ED Fig. 6a adds anything helpful.

Thank you for the suggestion. We will prepare a new figure that focuses on the gates to make this clearer. The comparison with Alphafold is valuable since the luminal loops and gates are not well modeled. Many groups are using these structures to do biochemical and computational experiments and perhaps even to design small-molecules. Since Alphafold differs substantially in this area, it might be of interest to those in the community doing this type of work.

The polar networks described in the manuscript provide interesting possibilities for interactions with substrates and protons whose binding to VMAT2 must control conformational change. Aside from the description of these networks, there is little evidence presented to assess the role of these networks in transport. Are the networks conserved in other closely related transporters? How could the interaction of the networks with substrate or protons affect conformational change? Of course, any potential role proposed for the networks would be highly speculative at this point, and any discussion of their role should point out their speculative nature and the need for experimental verification. Some speculation, however, can be useful for focusing the field's attention on future directions. However, statements in the abstract (three distinct polar networks... play a role in proton transduction.) and the discussion (...are likely also involved in mediating proton transduction.) should be clearly presented as speculation until they are validated experimentally.

We agree these statements are speculative, which we acknowledged in the text. We will further emphasize this point in a future revision. Please note, however, that many of these residues have been highlighted in other studies (30-32,37,49,55), and we have cited them in the text. Please see previous response.

Most of these residues are indeed highly conserved. It is a good idea to highlight this in our sequence alignment of related transporters. We will do so in our revised manuscript.

The strongest aspect of this work (aside from the structure itself) is the analysis of TBZ binding. There is a problematic aspect to this analysis. The discussion on how TBZ stabilizes the occluded conformation of VMAT2 is premature without structures of apo-VMAT2 and possibly structures with other ligands bound. We don't really know at this point whether VMAT2 might be in the same occluded conformation in the absence of TBZ. Any statements regarding the effect of interactions between VMAT2 and TBZ depend on demonstrating that TBZ has a conformational effect. The same applies to the discussion of the role of W318 on conformation and to the loops proposed to "occlude the luminal side of the transporter" (line 131).

Please see the response to this argument presented earlier. The occluded structure clearly shows the residues serving as gates. To understand how the gates open is a separate question. This does require additional structures and computations which are beyond the scope of this work. Our structure is interpreted in the context of all available biochemical data.

The description of VMAT2 mechanism makes many assumptions that are based on studies with other MFS transporters. Rather than stating these assumptions as fact (VMAT2 functions by alternating access...), it would be preferable to explain why a reader should believe these assumptions. In general, this discussion presents conclusions as established facts rather than proposals that need to be tested experimentally.

Indeed, the structural details of alternating access in MFS transporters are based on structures of other related proteins and we have cited review articles that describe this (29,30,48). We would like to highlight that these assumptions are not without merit, as previous studies investigating predicted gating residues (the same residues resolved in our structure) were based on studies of other MFS transporters and the demonstrated biochemical results are consistent with an alternating access transporter. These biochemical experiments also clearly demonstrate that a broadly similar mechanism of alternating access is used by VMAT2, see (30-32,48) which we have cited extensively when discussing these mechanisms.

The MD simulations are not described well enough for a general reader. What is the significance of the different runs? ED Fig. 4d is not high enough resolution to see the details.

We plan to provide additional experimental details and data to support the computational experiments in a revision. See response to reviewer #3.

Reviewer #3 (Public Review):

Summary:

The vesicular monoamine transporter is a key component in neuronal signaling and is implicated in diseases such as Parkinson's. Understanding of monoamine processing and our ability to target that process therapeutically has been to date provided by structural modeling and extensive biochemical studies. However, structural data is required to establish these findings more firmly.

Strengths:

Dalton et al resolved a structure of VMAT2 in the presence of an important inhibitor, tetrabenazine, with the protein in detergent micelles, using cryo-EM and with the aid of domains fused to its N- and C-terminal ends. The resolution of the maps allows clear assignment of the amino acids in the core of the protein. The structure is in good agreement with a wealth of experimental and structural prediction data and provides important insights into the binding site for tetrabenazine and selectivity relative to analogous compounds.

Weaknesses:

The authors follow up their structures with molecular dynamics simulations. The simulations resulted in repositioning of the ligand, which does not seem to be well founded, and raises questions about the methodological choices made for the simulations.

We appreciate the comments of reviewer #3 and thank them for these suggestions regarding the MD simulations. We will be supplying additional information to address the questions of reviewer #2 and #3 regarding the MD simulations including 1) movies which show there is not a substantial repositioning of ligand in any of the three runs 2) a table showing protonation states of residues and TBZ 3) data which shows that the number of waters which enter the binding site is relatively few compared with simulations of dopamine bound VMAT2 4) in run 2, more waters have entered the binding site vs. run 1 and 3 which likely explains why there is a small repositioning of TBZ.

We will also be providing a substantially improved map in a revised manuscript where the peripheral TMHs and loops are better resolved.

  1. Howard Hughes Medical Institute
  2. Wellcome Trust
  3. Max-Planck-Gesellschaft
  4. Knut and Alice Wallenberg Foundation