Small Molecule and Peptide Recognition of Protein Transmembrane

Mar 29, 2017 - The interactions of transmembrane domains are much less well understood than those of their water-soluble counterparts, and they have b...
0 downloads 0 Views 3MB Size
Subscriber access provided by UNIV OF REGINA

Current Topic/Perspective

Small molecule and peptide recognition of protein transmembrane domains Xianfeng Zeng, Peiyao Wu, Chengbo Yao, Jiaqi Liang, Shu-Ting Zhang, and HANG Hubert YIN Biochemistry, Just Accepted Manuscript • DOI: 10.1021/acs.biochem.6b00909 • Publication Date (Web): 29 Mar 2017 Downloaded from http://pubs.acs.org on March 30, 2017

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

Biochemistry is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 42

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biochemistry

Small molecule and peptide recognition of protein transmembrane domains Xianfeng Zeng,1 Peiyao Wu,1 Chengbo Yao,1 Jiaqi Liang,1 Shuting Zhang,1,3* Hang Yin1,2* 1

Center of Basic Molecular Science, Department of Chemistry, Tsinghua University, Beijing 100082 China. 2

Department of Chemistry and Biochemistry and BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80309, USA.

3

School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, China.

ACS Paragon Plus Environment

Biochemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Abstract: Membrane proteins play vital roles in cell signaling, molecular transportation and cell adhesion. The interactions of transmembrane domains are much less well understood than those of their water-soluble counterparts, and they have been deemed “undruggable” despite their important biological functions such as protein anchoring, signal transduction, and ligand recognition. Nevertheless, continual developments in this area have revealed useful probes for investigating and regulating these membrane proteins. This review summarizes and evaluates the strategies available for discovering small molecules and peptides that recognize the protein transmembrane domains of membrane proteins, with a particular focus on rational design and library screening.

ACS Paragon Plus Environment

Page 2 of 42

Page 3 of 42

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biochemistry

1. Introduction Membrane proteins (MPs) constitute about 23% of all human proteins and around 50% of membrane mass, and are involved in a huge number of crucial biological functions.1 Cell adhesion molecules such as integrins and selectins participate in cell communication between adjacent cells or between cells and the extracellular matrix, thus allowing cell migration and localization, which are key steps in oncogenesis and immune responses. Cell surface receptors such as epithelial growth factor receptors (EGFR), toll-like receptors (TLRs), and NOD-like receptors are able to distinguish molecule types and trigger downstream signaling pathways. For instance, EGFR undergoes a transition from an inactive monomeric form to an active homodimer upon EGF binding, and subsequently initiates downstream gene activation and signaling pathways. TLRs and NOD-like receptors which belong to the pattern recognition receptor family and trigger immune responses via the identification of structurally conserved molecules from microbes or stressed cells are termed as pathogen-associated molecular patterns and damage-associated molecular patterns, respectively. Membrane transporter proteins including carriers and channels are crucial for cell growth, homeostasis and the conduction of nerve pulses due to their ability to facilitate materials across the cellular membrane. Large groups of membrane-bound enzymes are also involved in various vital metabolic reactions. Therefore, MPs are critical targets in drug discovery due to their indispensability in biological functions and their dysregulation leads to severe cellular malfunctions and diseases. Transmembrane domains (TMDs) are fundamental in anchoring the MPs to the cellular surface, and the subsequent oligomerization is important in signal transduction and ligand recognition.2 Due to their crucial cell signaling transduction abilities, MPs make up approximately 60% of drug targets. However, targeting these TMDs with exogenous agents still remains a daunting challenge due to several reasons.3 First, conventional probes (e.g., antibodies) are implicated with loss of function within phospholipid bilayers due to the highly hydrophobic environment. Second, an appropriate level of hydrophobicity is required for the probes to properly interact with the lipid bilayer. Other properties such as hydrogen bonds, electrostatic interactions, and polarpolar interactions are also essential for TMD target selectivity and high affinity. Lastly, the binding of TMDs

ACS Paragon Plus Environment

Biochemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 4 of 42

rarely takes place in distinguished pockets such as enzymes, making them difficult for small molecules to seek out. Despite these major hurdles, recent advances have been made in the discovery of TMD probes. Herein, we introduce two general strategies for the discovery of TMD targeting agents, focusing specifically on rational design and library screening (Figure 1).

Figure 1. Overviews of major methods to identify agents recognizing MP TMDs.

2. Rational design of agents targeting TMDs 2.1 Rational design based on natural product scaffolds Nature offers inspiration for rational drug design with bioactive molecules. Novel probes with a higher affinities or extended functions are obtained by modifying the structure of natural TMD-recognizing agents (Figure 2). While this approach has been fruitful, the main issue needed to be considered is that modification of natural scaffolds may affect the interactions between the molecule and its MP target.

ACS Paragon Plus Environment

Page 5 of 42

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biochemistry

Figure 2. Rational design approaches based on natural product scaffolds. One of the approaches for the discovery of new agents recognizing protein TMDs is to attach a new pharmacophore to natural small molecules (left). The high affinity between nature-existed molecules and their TMD targets would guide the new attached pharmacophore to the binding sites; Also, structural modifications on natural small molecules could improve the affinity between those natural product analogs and TMD target (right).

Natural products are first modified to obtain probes designed to specifically target various TMD conformations. The discovery of specific bile acid receptor agonists exemplifies this kind of modification. Bile acids including cholic acid and lithocholic acid (LCA) are amphipathic steroid acids. They are recognized by nuclear bile acid receptors, such as the Farnesoid X receptor (FXR) and G protein-coupled bile acid receptor (GPBAR1, also known as TGR5). Insufficient bile acid signaling leads to adverse metabolic disorders such as cholesterol maladies and type II diabetes.4 Various modifications of bile acids have been explored to obtain drugs that can selectively activate one of the two receptors to reduce clinical side effects. Francesco et al. explored interactions between bile acid derivatives and human TGR5, and identified 3-amino-LCA derivatives 1-3 (Figure 3), a series of TGR5-specific agonists with low EC50 values. Homology modeling and in silico simulations predicted that the Glu169 anion within the TMD of hTGR5 interacts with the 3α-hydroxyl group in the LCA scaffold via hydrogen bonding, whereas in FXR, the positively charged His444 within the TMD interacts with the oxygen atom in 3α-hydroxyl in the LCA scaffold. Consequently, an ammonium cation was introduced at the C3 position of the LCA by replacing a primary amino group with a hydroxyl group, which successfully reduced its affinity toward FXR.5

ACS Paragon Plus Environment

Biochemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 6 of 42

Figure 3. The structure of selective TGR5 agonists 1-3. Compounds 1-3 were obtained by replacing hydroxyl group with amino group in scaffold of LCA.

Linking various pharmacophores to the natural substrates of transporters is another common strategy for drug development. For example, a hexose-conjugate, with a combination of hexose moiety and anticancer pharmacophore, is selective toward cancer cells that recognize glucose transporters (GLUTs).6 GLUTs are vital in facilitating the transportation of glucose and other carbohydrates across cell membranes, a process that is especially important in cancer cells. Cancer cells generally show a low metabolic efficiency under anaerobic conditions, and require an increased level of hexose uptake to enable their unregulated growth (a.k.a. Warburg effect, Figure 4A).7, 8 As a consequence, high levels of hexose uptake and GLUTs expression are indicative of several types of cancer.9-11 Patra et al. designed three glucose-platinum conjugates that show great cytotoxicity toward cancer cells. Based on the crystal structure of XylE, a GLUT1 homolog, they conjectured that the hydroxyl group on the C6 of glucose is not an active participant in the glucose-GLUT recognition, and modification of this hydroxyl group might be tolerated in transporter recognition. The results of docking designed conjugates (Glc-Pts 4-6 Figure 4C) into XylE show that these conjugates form hydrogen bonds with various residues in XylE TMD. Further biochemical experiments indicated that these conjugates selectively accumulated in cancer cells (ovarian cancer cells A2780 and prostate cancer cells DU145) while having little impact on noncancerous neuron cells in vitro (Table 1).12 Similarly, Wu et al. reported galactose conjugated cisplatinum (Glc-Pts 7, Figure 4C) with promising efficacy against lung cancer and colon cancer.13

ACS Paragon Plus Environment

Page 7 of 42

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biochemistry

Figure 4. Warburg effect and strategies of developing small-molecule drugs exploiting Warburg effect. (A) Warburg effect: GLUTs are overexpressed in cancer cells to compensate for low metabolic efficiency, and enable unregulated cell growth; (B) Strategies exploiting Warburg effect: Glucose conjugates enable selective delivery of anticancer pharmacophores to the cancer cells (left); GLUTs inhibitors can directly block the glucose uptake in energy demanding cancer cells (right); (C) The structure of designed hexose conjugates and their aglycone moiety.

ACS Paragon Plus Environment

Biochemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 8 of 42

Table 1. Effects of Glc-Pts 4 and its aglycone moiety on the platinum uptake and reduction of cell viability in ovarian cancer cell line A2780 and prostate cancer cell line DU145. Uptake (pmol Pt/106 cells)

Reduction in cell viability (%)

A2780

DU145

A2780

DU145

4

19.0±0.7

116.6±4.3

44.7±1.2

58±3.5

aglycone

9.4±0.5

69±0.09

16.6±1.7

35±3.2

Certain antibiotics and cytotoxins regulate activities of enzyme and transporter via specific interactions of membrane-bound TMD.14 The identification of antibiotic and TMD interactions, followed by investigation of the structure-activity relationship (SAR) among the modified antibiotics, can be a useful strategy for overcoming multidrug resistance in certain types of bacteria. Bacterial lipoprotein signal peptidase II (LspA) is a key enzyme in the post-translational lipidation of bacterial proteins, and plays an important role in controlling the physiology and pathogenicity of bacteria.15 Globomycin 8 (Figure 5A), a 19-membered cyclic depsipeptidic antibiotic isolated from Streptomyces halstedii,16 can inhibit Gram-negative bacteria growth by interacting with LspA.17 Further research reveals that the disruption of the normal post-translational lipidation by 8 leads to the accumulation of diacylglyceride-containing lipoprotein precursors in the cytoplasmic membrane,18 which is toxic to bacteria. Kiho et al. conducted the SAR study of 8 and showed that the length of the alkyl side chains and the hydroxyl group in L-Serine are both essential for inhibitory activity.19 The alkyl chain modification resulted in an analog 9 (Figure 5A) that was 10 times more potent than 8. In addition, compound 9 showed a wider antibacterial spectrum than 8 (Table 2). This study exemplifies the viability of modifying antibiotics to counteract drug resistance. Vogeley et al. solved the crystal structure of the LspA complex from Pseudomonas aeruginosa with 8 at 2.8 Å resolution (Figure 5B).20 Using molecular dynamics simulation, they proposed that 8 interacted with LspA via hydrophobic and hydrogen bonding interactions with certain Asp residues (Asp124 and Asp143) that are responsible for catalyzing the signal peptide hydrolysis. The authors also pointed out that modification of the alkyl chain could strengthen the interactions between 8 analogs and the apolar surface of the

ACS Paragon Plus Environment

Page 9 of 42

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biochemistry

MH2 transmembrane helix, leading to a higher affinity with analog 9.20 These results pave the way for the design of antibacterial drugs targeting LspA.

Figure 5. The molecular structure of globomycin and its co-crystal structure of globomycin in complex with LspA. (A) The structure of globomycin (8) and its analog 9; (B) The crystal structure of LspA-globomycin complex. (PDB entry: 5DIR). Modified from Reference 20. Figure 5B is reprinted with permission.

Table 2. Antimicrobial activity of globomycin (8) and its analog 9 against E. coli and S. aureus (MRSA). Minimum inhibitory concentration (MIC, ug/mL) E. coli

S. aureus (MRSA)

8

12.5

>100

9

1.56

12.5

2.2 Truncated peptides Interactions between TMDs contribute to the overall process of MP oligomerization. ToxR assay is generally applied to investigate the interactions between TMDs (Figure 6A). ToxR, derived from Vibrio cholerae, is a single-pass transmembrane activator located on the inner membranes of bacteria. Dimeric ToxR activates the ctx promoter and subsequently triggers reporter gene expression. By combining ToxR with various TMDs, the associations of TMDs can be evaluated by the expression level of the reporter gene. Langosch et al. first proposed ToxR assay and demonstrated that the GXXXG motif of GpA TMD was critical in its dimerization.21, 22

Similarly, Russ et al. used an alternative reporter, chloramphenicol acetyltransferase (CAT), and developed

the TOXCAT assay.23 Other reporter gene systems, such as lacZ and firefly luciferase, have also been applied to ACS Paragon Plus Environment

Biochemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 10 of 42

ToxR systems.24 Dominant-negative TOXCAT (DN-TOXCAT), a variation of TOXCAT technique, is used to measure the heterodimerization of TMDs. One TMD is fused with a functional ToxR while a second TMD is fused with a mutant ToxR. Because only the dimer formed by two functional ToxR initiates a downstream CAT synthesis, the subsequent CAT signal correlates with the strength of TMD interaction. Although TOXCAT was originally designed to study single pass TMD dimerization, Yin and co-workers showed that it is also useful in investigating interactions of multipass transmembrane helices.25

Figure 6. TOXCAT system and truncated peptides as probes recognizing MP TMDs. (A) TOXCAT system: MBP: maltose binding protein; TM1 and TM2: Two engineered transmembrane helices; TM1 and TM2 interact with each other and form a functional ToxR dimer. The ToxR dimer binds to ctx promoter and induces measurable synthesis of CAT. TOXCAT provides a general way to determine the heterodimerization of two transmembrane helices; (B) Truncated peptides: Once the interacting protein transmembrane helices are determined, the truncated TMDs of one oligo partner can be exploited to modulate native oligomerization.

ACS Paragon Plus Environment

Page 11 of 42

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biochemistry

Naturally existing interactions among TMD helices within MP oligomers can be exploited to develop truncated TMD peptides and to regulate the activities of the original MPs (Figure 6B). This approach, known as truncated peptides, allows discovery of peptides regulating MP oligomerization process, which has resulted in several useful probes for TLRs, insulin receptors (IRs), and Her (ErbB) family proteins. Compared to small-molecular probes, truncated peptides are selective and efficacious with less off-target effects. TLRs play critical roles in the innate immune system by specifically recognizing a variety of pathogen- and damage-associated molecular patterns.26 Among them, the heterodimerization between TLR2 and TLR1 or TLR6 is crucial for host immunity against lipopeptides anchored on the bacterial surface.27, 28 Using the ToxR assay, Godfroy et al. demonstrated the dimerization of various TLRs and their subsequent signaling transduction.29 Based on these studies, Fink et al. took advantage of the truncated TMDs of TLR2 and TLR6 to inhibit their dimerization and obstruct the subsequent signaling pathway.30 Due to the resemblance between TLR1 TMD and TLR6 TMD, both of these truncated TMD peptides are able to antagonize the secretion of proinflammatory cytokines such as TNF-α and interleukin 6 (IL-6) in macrophages after treatment of lipoteichoic acid (LTA, TLR 2/6 agonist) and Pam3CSK4 (TLR1/2 agonist), but not lipopolysaccharide (LPS, TLR4 agonist). Comprehensive biochemical and biophysical experiments have verified the interactions between these truncated peptides and their reciprocal TLRs. In vivo studies showed that truncated peptides derived from TLR1 and TLR6 were able to rescue mice with TLR2-related fatal inflammation, but not TLR4.29 Moreover, Shmuel-Galia et al. utilized this TLR2 truncated peptide to further investigate the role of TLR2 heterodimerization in the activation of pathogenic pro-inflammatory Ly6Chi monocytes and demonstrated the therapeutic potential of truncated TMD peptides in ameliorating colitis.31 Shoelson and co-workers applied this strategy of using TM domain sequence peptide to identify agonist of IR.32 IR is a member of receptor tyrosine kinase (RTK) family, and plays an essential role in cell metabolism and growth.33-35 Shoelson et al demonstrated that a 24-residue IR TMD containing oligopeptide can specifically activate IR signaling. Based on biochemical analyses, the dissociation of two inactive IR TMD dimers potentially leads to closer proximity of two β-subunits and initiates the IR activation.36, 37 Upon the binding of truncated IR-TMD peptide, the dimerization of two inactive IR TMDs collapses, resulting in the juxtaposition of the two β-subunits and IR auto-phosphorylation. ACS Paragon Plus Environment

Biochemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 12 of 42

Due to their great importance in human cancer pathogenesis, the TMDs of the ErbB family proteins have been thoroughly studied.38-40 Truncated peptidic probes have also been involved in the study of the Her (ErbB) family of RTKs including ErbB1 (Her1/ EGFR), ErbB2 (Her2/ Neu), ErbB3, and ErbB4. ErbB receptor family has essential roles in mammalian development and homeostasis, and thus these receptors are considered as promising targets for anticancer drugs. 41, 42 Structural studies implicated the role of EGFR dimerization in the activation of cytoplasmic protein tyrosine kinase activity.43 Lemmon and co-workers demonstrated via TOXCAT assay that the C-terminal GXXXG motif was involved in homodimerization in ErbB proteins.44 Shai’s group revealed that the N-terminal GXXXG motif participates in the heterodimerization between ErbB1 and ErbB2.45 This conclusion was further verified by the ability of a synthetic ErbB1 transmembrane peptide to heterodimerize with ErbB2. Subsequent studies showed that the TMD peptide of ErbB1 and ErbB2 could specifically inhibit the auto-phosphorylation and subsequent signal transduction of their cognate receptors.46

2.3 Optical isomers of transmembrane helices Peptides composed of D-amino acids may retain their ability to interact with the binding partners of their naturally occurring enantiomers. Meanwhile, these peptides tend to be resistant to enzyme degradation, thus show desirable stability as potential anti-TMD probes (Figure 7).

ACS Paragon Plus Environment

Page 13 of 42

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biochemistry

Figure 7. Targeting the TMDs with optical isomers. D-enantiomers possess same amino acid sequence but different residue orientations comparing with their L-counterpart, by changing the tilt angle between two helices, it is possible to bring the similar interacting surfaces orientation as the L-homodimer did and with some special stability and affinity to their TMD target.

This strategy has been successfully used to develop potential drugs against human immunodeficiency virus (HIV-1) infection. The N-terminal domain of HIV-1 glycoprotein 41 (gp41) was demonstrated to be involved in the infection of the host cell.47 Kliger et al. synthesized a 33-residue oligopeptide corresponding to the Nterminal domain of gp41, which showed inhibition of cell fusion through interaction with the N-terminus of gp41.48 ATR-FTIR spectroscopy demonstrated that two enantiomers have a similar secondary structure in lipid bilayers, suggesting that highly hydrophobic environment within TMDs may help to hide backbone polar groups and maintain the enantiomers’ secondary structure. Based on this result, Pritsker et.al showed that an all Damino acid peptide (D-WT) in line with the N-terminal domain of gp41 inhibited cell fusion through gp41.49 Interestingly, D-WT did not inhibit cell fusion mediated by the HIV-2 envelope glycoprotein.

ACS Paragon Plus Environment

Biochemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 14 of 42

Shai’s group synthesized D-amino acid peptides to target TMDs of glycophorin A (GPA) and T-cell receptor (TCR) complex. The synthesized all-D analog of GPA TMD was showed to form dimmer with native GPA TMD.50 In an alternative approach, a diastereomer of GPA TMD by replacing two L-valines with D-valines was synthesized. The designed peptide exhibited a dose-dependent dominant negative effect on the dimerization of GPA TMD in ToxR assay. Molecular dynamics showed that both the homodimer and heterodimer maintained a similar contact surface, which could explain why the D-GPA retained the ability to bind to L-GPA.51 Shai and co-workers also developed a D-peptidic probe inhibiting T-cell activation based on the peptide previously reported to interfere with the interaction between the TCR α-chain and the cluster of differentiation 3 (CD3) Tcell co-receptor.52 Similarly, both the all-D and the diastereomer analogs with an L-arginine residue or an Llysine residue were used to inhibit antigen-specific T-cell activation and adjuvant arthritis in vivo in rats. Importantly, compared with all-L forms, the diastereomer analogs showed greater immunosuppressive activity in vivo and in vitro, which could be attribute to its greater solubility and resistance to degradation.53, 54 In general, truncated peptides and optical isomers can be regarded as another type of modification on natural MP oligomer scaffolds. Similar to modification on small molecules, the success of such designed truncated peptides (or optical isomers) depends critically on the ability of modified peptides to retain the native interactions with their oligomer partners. With the help of molecular dynamics and MP structural information, peptide design is expected to become a more efficient and reliable method in modulating MP oligomers.

2.4 CHAMP peptides With the rapid development of technology, computational design has become an increasingly feasible method for the development of peptides targeting protein TMDs. Yin et al. developed a computer-based strategy, computed helical anti-membrane protein (CHAMP), to identify peptides that could sequence-specifically interact with TMD helices (Figure 8).55 The initial step in the development of the CHAMP peptidic probe involved the identification of a preferred TMD dimerization mode between the target and the CHAMP anti-TMD peptide based on the structures in the known TMD database. After threading the target sequence into one of the TMD helices, the CHAMP’s sequence was optimized using a Monte Carlo repacking algorithm. Finally, the noninteracting residues were selected for insertion and anchoring with an Ez potential model.56 The CHAMP ACS Paragon Plus Environment

Page 15 of 42

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biochemistry

method was used to study two homologous integrins, αIIbβ3 and αVβ3, the activation of which is regulated by the heterodimeric TMD association. The activities of the designed CHAMP peptide in micelles, liposomes and mammalian cells were also confirmed in the study.55 In addition, Caputo et al. showed the direct interaction between the CHAMP peptide and the full-length αIIbβ3.57

Figure 8. Design of CHAMP peptides targeting TMDs. (A) The general procedure to predict CHAMP peptide interface: The first step is to select a backbone geometry from the known structures that contain motifs in TMD target. Then amino acid residues are added to backbone, followed by a side chain-repacking algorithm; (B) The sequence motifs of αIIbβ3 and αVβ3; (C) The predicted packing interface between the integrin and CHAMP; (D) Activation of integrins by CHAMP peptide. Modified from Reference 55. Figure 8 is reprinted with permission.

Natural peptides consist of α-amino acids, while Kritzer et al. designed β-peptides that were made of β-amino acids to inhibit protein-protein interactions.58 However, the computational design of β-peptides is more challenging due to the lack of appropriate force fields. Nonetheless, Shandler et al. successfully applied the CHAMP method to design a β-peptide that targeted the transmembrane helix of the integrin αIIbβ3.59 The secondary structure and orientation of the peptide in the phospholipid bilayer were characterized using circular dichroism spectroscopy and attenuated total internal reflectance IR spectroscopy. Furthermore, it was confirmed that the designed peptide could selectively interact with the isolated TMD of αIIbβ3 and activate the integrin in vitro by several other techniques including analytical ultracentrifugation, rupture force spectroscopy, and transmission electron microscopy. 59

ACS Paragon Plus Environment

Biochemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 16 of 42

3. Screening of agents targeting TMDs 3.1 Traptamers Transmembrane peptide aptamers, also known as traptamers, are artificial transmembrane peptides that are selected for targeting specific TMD of MPs (Figure 9). The bovine papillomavirus E5 protein, a 44 amino acid oligopeptide, is the smallest oncoprotein.60 E5 transforms cells by interacting with the TMD of platelet-derived growth factor β receptor (PDGFβR) to initiate dimerization and trans-autophosphorylation.61 DiMaio’s group conjectured that the E5 protein could serve as a scaffold to facilitate the discovery of probes targeting other cellular MPs. They constructed a traptamer peptide library by replacing E5 TMD with random hydrophobic amino acid sequences. This library was successfully applied to screen for TMD peptides that recognize the human erythropoietin receptor (EpoR). An artificial oligopeptide that activated human EpoR (without affecting murine EpoR and PDGFβR) was identified.62 This result indicated that the sequence changes within the TMD of a viral oncogene product could benefit the recognition of a completely unrelated protein target. Using a similar strategy, the group developed traptamers to identify six 44 or 45 amino acid oligopeptides that recognized the TMD of CCR5, a co-receptor of human HIV.63

ACS Paragon Plus Environment

Page 17 of 42

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biochemistry

Figure 9. Targeting the TMDs by traptmers. Traptmer library can be constructed by replacing the TMD of E5 peptide with hydrophobic residues. High affinity traptmers for the target MP TMD will be identified by genetic screening.

3.2 In silico screening High-throughput screening, which involves rapid screening of compound libraries, may provide a reasonable alternative to rational design when the molecular basis is unclear.2 Various screening methods have been used in discover molecules targeting MPs, including wet screening and in silico screening. Compared to wet screening, in silico screening allows a more cost-efficient approach to identify hit molecules from a large library, which has been widely used to identify small molecules targeting G-protein coupled receptor (GPCR), GLUTs and many other MP targets.

ACS Paragon Plus Environment

Biochemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 18 of 42

A prominent example of in silico screening to identify novel GPCR ligands is PZM21, a selective µ-opioidreceptor (µOR) agonist with little side effects. Morphine is a widely used canonical opioid analgesic that exerts its effect by activating µOR signaling.64 However, the side-effects of severe respiratory depression caused by the activation of µOR downstream in the β-arrestin pathway, and other closely related receptors, limit its analgesic application.65-67 Manglik et al. docked over 3 million compounds from the ZINC database against the inactive form of the µOR structure. Structure-based optimization on high-affinity hits yielded PZM21 (IC50=1 nM), with a 1000-fold improvement in binding infinity. PZM21 shared no structural similarity with canonical opioid drugs, and exhibited different in vivo profiles, as it only modulated brain µOR with no obvious effect on spinal cord µOR. In vivo study demonstrated that PZM21 had high analgesic efficacy, and was less likely to produce respiratory depression and constipation side effects, which were some of the major limitations of canonical opioid agonists.68 This work highlighted structure-based screening and its ability to identify GPCR ligands with new chemotypes that offer new efficacy profiles. However, the authors pointed out that screening small molecules followed by extensive SAR studies on one subtype target to acquire high selectivity might not be applicable in all situations.68 Orphan receptors are formidable targets for drug discovery, mainly due to the difficulties involved in developing functional assays without known ligands.69 Huang et al. described a combined in silico screening and in vitro approach to study the ligands of these pharmacologically dark receptors.70 A non-selective GPR68 agonist, lorazepam, was first identified. Through iterative modeling and optimization of 3307 homology models, a putative binding mode of lorazepam-GPR68 was obtained, and further verified by site-mutation of the putative interacting residues within TMDs. Based on this optimized model, ogerin (a selective GPR68 ligand) was identified. Ogerin was suggested to interact with Glu180, a residue located in the TMD and was not involved in the lorazepam-GPR68 interaction, which contributed to the isoform selectivity. Using ogerin as a probe, some important biological functions of GPR68 were revealed in the mouse hippocampus. Two molecules that potentially bind to GPR65 were identified using the same method, which indicated its general applicability.70 Developing GLUTs inhibitors that directly block the uptake of glucose in cancer cells is another strategy that exploits the Warburg effect against cancers. The crystal structures of GLUT1,71 GLUT3,72 GLUT573 reveal that the GLUT family proteins have similar structures, with promising potential pockets for the development of ACS Paragon Plus Environment

Page 19 of 42

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biochemistry

inhibitors. Thompson et al. performed virtual screening on an inward-open GLUT5 homology model and selected the 175 top-ranked compounds for in vitro tests.74 The hit compound, MSNBA, selectively inhibited GLUT5 without affecting GLUT1-4 activity. The docking and mutagenesis results suggested that the interaction between the amino group in MSNBA and His387 on GLUT5 determined the specificity of MSNBA towards GLUT5. This work implied that the subtle differences between the crystal structures of GLUTs could be exploited to develop selective inhibitors. Mishra et al. reported two GLUT4 selective inhibitors using in silico screening.75 They constructed a GLUT4 homology model and virtually screened 1.8 million compounds from the ZINC database. A cell-viability assay determined that two hit compounds were effective. Two hit compounds were observed to specifically inhibit GLUT4 (not GLUT1) mediated glucose uptake in HEK293 cells that exogenously express GLUT1 and GLUT4. The results of the docking the two hits to the GLUT1 crystal structure and GLUT4 homology model revealed the interacting residues that contribute to selectivity. This study highlighted the power of homology modeling in acquiring GLUT isoform selectivity. Whereas it is still challenging to accurately predict the molecular recognition toward different MP conformations and assess the off-target effects using in silico screening. With accumulating MP structural information and improved computational algorithm, in silico screening will continue to be an attractive strategy in identifying small molecules recognizing MP TMDs.

3.3 Wet screening Although examples are rare, wet screening can also serve as a powerful tool to identify small molecules regulating vital processes within TMDs. M. tuberculosis (MTB) is the causative agent of most cases of tuberculosis (TB). MTB can turn into a dormant state, thus nullifying the potency of many common antibiotics. Due to its vital role in energy metabolism that is required by both active and dormant bacteria, MTB ATPase has become an ideal drug target in the development of new anti-TB drugs.76 In 2005, Andries et al. identified a diarylquinoline-based drug, TMC207, also known as bedaquiline, using a whole-cell assay to inhibit multiple growth cycles of TB.77 Analysis of the resistant mutants identified TMC207, which acted on the TMD of the F0 subunit of mycobacterial ATP synthase. By blocking the ATPase rotation motor, TMC207 was able to inhibit both dormant and metabolizing cells.77 Biukovic et al. reported the structure of the MTB F1F0 ATP synthase ε ACS Paragon Plus Environment

Biochemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 20 of 42

subunit and C-terminal segment, and presented a new model of the TMC207-ATPase binding mode. It was suggested that wedge-like TMC207 interacted with Trp15 and Phe50 in ring c and subunit ε, hindering the function of ring c’s rotation and subunit ε’s regulation of coupling, respectively.78 These studies offered more detailed information about the TMC207-ATPase binding mode, which led to the further discovery of TMC207 analogs that target resistant strains. In 2012, the FDA approved TMC207 for treating multidrug resistant TB.79 There are very few examples of using small molecule agents to target lateral transmembrane protein-protein interaction. Human herpes virus 4 (EBV) is a common virus in humans, and is related to several autoimmune diseases80 and B cell lymphomas.81 The latent membrane protein 1 (LMP-1) is an important oncoprotein involved in EBV infection. LPM-1 TMD5 forms a homo-trimeric complex to initiate constitutive signaling and immortalize B cells during EBV infection. Through alanine scanning, Yin and co-workers discovered that the fifth transmembrane helix of LMP-1 was responsible for homotrimerization. In addition, a mutation of Asp150 on the fifth transmembrane helix of LMP-1 abolished the expression of NF-κB, a downstream signaling factor of LMP-1. This residue was involved in the hydrogen bond network, which stabilizes the formation of LMP-1 oligomers.82 Yin and co-workers screened the NCI library, and identified an inhibitor (NSC 295242) to disrupt the LMP-1 TM5 interactions. NSC 295242 was found bound to the hot spot on TM5, and thus inhibited LMP-1 homotrimerization.83

4. Perspective Technological progress has made many previously undruggable targets feasible for therapeutic development. Nonetheless, the regulation of TMDs by small molecules or peptides is still considered a formidable task. Despite the great difficulty involved in the discovery of probes against TMDs, recent developments in both library screening and rational design have illuminated this obscure area. High-throughput screening, either target-based or phenotype-based, followed by an extensive SAR study, is always a useful approach to identify lead compounds. Aided by computational design together with improved structural knowledge obtained by cryo-EM, X-ray crystallography, top-down MS, and molecular dynamics, researchers have developed new procedures to obtain hits toward various MPs. In silico screening is becoming

ACS Paragon Plus Environment

Page 21 of 42

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biochemistry

de rigueur for the discovery of lead compounds for TMDs, especially GPCRs. We expect that with more precise energy functions, in silico screening will become more accurate and efficient. As an alternative way to generate probes, rational design enables researchers to modify existing probes to improve their potency and stability, based on a comprehensive understanding of the underlying interaction modes. Rational design depends on a comprehensive understanding of MPs. The already discovered probes are a treasure trove, inspiring more novel and fascinating strategies for drugging the currently undruggable MP TMDs.

ACS Paragon Plus Environment

Biochemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 22 of 42

5. Author Information Corresponding author *Email: [email protected] (H. Y.); [email protected] (S.Z.)

Funding Funding of this work was provided by National Natural Science Foundation of China (NSFC Grant No. 21572114) and Tsinghua University.

Notes The author declares no competing financial interest.

ACS Paragon Plus Environment

Page 23 of 42

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biochemistry

6. Acknowledgements We thank the other group members for their comments and assistance on this work.

ACS Paragon Plus Environment

Biochemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 24 of 42

7. Abbreviations MPs, membrane proteins; EGFR, epithelial growth factor receptor; TLRs, toll-like receptors; PRR, pattern recognition receptor; TMDs, transmembrane domains; LCA, lithocholic acid; FXR, Farnesoid X receptor; GLUTs, glucose transporters; Glc-Pts, glucose-platinum conjugates; Gal-Pts, galactose platinum conjugates; SAR, structure-activity relationship; LspA, lipoprotein signal peptidase II; CAT, chloramphenicol acetyltransferase; DN-TOXCAT, dominant-negative TOXCAT; IR, insulin receptor; LTA, lipoteichoic acid; LPS, lipopolysaccharide; RTK, receptor tyrosine kinase; gp41, HIV-1 glycoprotein 41; D-WT, D-amino acid peptide; GPA, glycophorin A; TCR, T-cell receptor; CD3, cluster of differentiation 3; CHAMP, computed helical anti-membrane protein; GPCR, G protein-coupled receptor; µOR, µ-opioid-receptor; PDGFβR, plateletderived growth factor β receptor; EpoR, erythropoietin receptor; MTB, M.tuberculosis; TB, tuberculosis; EBV, human herpesvirus 4; LMP-1, latent membrane protein 1.

ACS Paragon Plus Environment

Page 25 of 42

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biochemistry

8. References [1] Uhlen, M., Fagerberg, L., Hallstrom, B. M., Lindskog, C., Oksvold, P., Mardinoglu, A., et al. (2015) Tissuebased map of the human proteome. Science 347, 1260419. [2] Yin, H., and Flynn, A. D. (2016) Drugging membrane protein interactions Annu. Rev. Biomed. Eng. 18, 5176. [3] Overington, J. P., Al-Lazikani, B., and Hopkins, A. L. (2006) Opinion - How many drug targets are there? Nat. Rev. Drug Discov. 5, 993-996. [4] Fiorucci, S., Mencarelli, A., Palladino, G., and Cipriani, S. (2009) Bile-acid-activated receptors: targeting TGR5 and farnesoid-X-receptor in lipid and glucose disorders. Trends Pharmacol. Sci. 30, 570-580. [5] Di Leva, F. S., Festa, C., Renga, B., Sepe, V., Novellino, E., Fiorucci, S., et al. (2015) Structure-based drug design targeting the cell membrane receptor GPBAR1: exploiting the bile acid scaffold towards selective agonism. Sci. Rep. 5, 8. [6] Calvaresi, E. C., and Hergenrother, P. J. (2013) Glucose conjugation for the specific targeting and treatment of cancer. Chem. Sci. 4, 2319-2333. [7] Warburg, O. (1956) Origin of cancer cells. Science 123, 309-314. [8] Vander Heiden, M. G., Cantley, L. C., and Thompson, C. B. (2009) Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324, 1029-1033. [9] Amann, T., and Hellerbrand, C. (2009) GLUT1 as a therapeutic target in hepatocellular carcinoma. Expert Opin. Ther. Targets 13, 1411-1427. [10] Shim, B. Y., Jung, J. H., Lee, K. M., Kim, H. J., Hong, S. H., Kim, S. H., et al. (2013) Glucose transporter 1 (GLUT1) of anaerobic glycolysis as predictive and prognostic values in neoadjuvant chemoradiotherapy and laparoscopic surgery for locally advanced rectal cancer. Int. J. Colorectal Dis. 28, 375-383. [11] Ramani, P., Headford, A., and May, M. T. (2013) GLUT1 protein expression correlates with unfavourable histologic category and high risk in patients with neuroblastic tumours. Virchows Arch. 462, 203-209. [12] Patra, M., Johnstone, T. C., Suntharalingam, K., and Lippard, S. J. (2016) A potent glucose-platinum conjugate exploits glucose transporters and preferentially accumulates in cancer cells. Angew. Chem. Int. Edit. 55, 2550-2554. ACS Paragon Plus Environment

Biochemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 26 of 42

[13] Wu, M., Li, H., Liu, R., Gao, X. Q., Zhang, M. H., Liu, P. X., et al. (2016) Galactose conjugated platinum(II) complex targeting the Warburg effect for treatment of non-small cell lung cancer and colon cancer. Eur. J. Med. Chem. 110, 32-42. [14] Klepsch, F., Jabeen, I., Chiba, P., and Ecker, G. F. (2010) Pharmacoinformatic approaches to design natural product type ligands of ABC-Transporters. Curr. Pharm. Design 16, 1742-1752. [15] Sankaran, K., and Wu, H. C. (1994) Lipid modification of bacterial prolipoprotein-transfer of diacylglyceryl moiety from phosphatidylglycerol. J. Biol. Chem. 269, 19701-19706. [16] Inukai, M., Nakajima, M., Osawa, M., Haneishi, T., and Arai, M. (1978) Globomycin- new peptide antibiotic with spheroplast-forming activity. II. Isolation and physicochemical and biological characterization. J. Antibiot. 31, 421-425. [17] Inukai, M., Takeuchi, M., Shimizu, K., and Arai, M. (1978) Mechanism of action of globomycin. J. Antibiot. 31, 1203-1205. [18] Hussain, M., Ichihara, S., and Mizushima, S. (1980) Accumlation of glyceride-containing precursor of the outer-membrane lipoprotein in the cytoplasmic membrane of escherichia-coli treated with globomycin J. Biol. Chem. 255, 3707-3712. [19] Kiho, T., Nakayama, M., Yasuda, K., Miyakoshi, S., Inukai, M., and Kogen, H. (2004) Structure-activity relationships of globomycin analogues as antibiotics. Bioorg. Med. Chem. 12, 337-361. [20] Vogeley, L., El Arnaout, T., Bailey, J., Stansfeld, P. J., Boland, C., and Caffrey, M. (2016) Structural basis of lipoprotein signal peptidase II action and inhibition by the antibiotic globomycin. Science 351, 876-880. [21] Langosch, D., Brosig, B., Kolmar, H., and Fritz, H. J. (1996) Dimerisation of the glycophorin a transmembrane segment in membranes probed with the ToxR transcription activator. J. Mol. Biol. 263, 525-530. [22] Brosig, B., and Langosch, D. (1998) The dimerization motif of the glycophorin A transmembrane segment in membranes: Importance of glycine residues. Protein Sci. 7, 1052-1056. [23] Russ, W. P., and Engelman, D. M. (1999) TOXCAT: A measure of transmembrane helix association in a biological membrane. Proc. Natl. Acad. Sci. U. S. A. 96, 863-868. [24] Roth, L., Nasarre, C., Dirrig-Grosch, S., Aunis, D., Cremel, G., Hubert, P., et al. (2008) Transmembrane domain interactions control biological functions of neuropilin-1. Mol. Biol. Cell 19, 646-654. ACS Paragon Plus Environment

Page 27 of 42

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biochemistry

[25] Joce, C., Wiener, A. A., and Yin, H. (2011) Multi-Tox: Application of the ToxR-transcriptional reporter assay to the study of multi-pass protein transmembrane domain oligomerization. Biochim. Biophys. ActaBiomembr. 1808, 2948-2953. [26] Kawai, T., and Akira, S. (2010) The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat. Immunol. 11, 373-384. [27] Jin, M. S., Kim, S. E., Heo, J. Y., Lee, M. E., Kim, H. M., Paik, S. G., et al. (2007) Crystal structure of the TLR1-TLR2 heterodimer induced by binding of a tri-acylated lipopeptide. Cell 130, 1071-1082. [28] Kang, J. Y., Nan, X., Jin, M. S., Youn, S. J., Ryu, Y. H., Mah, S., et al. (2009) Recognition of lipopeptide patterns by Toll-like receptor 2-Toll-like receptor 6 heterodimer. Immunity 31, 873-884. [29] Godfroy, J. I., Roostan, M., Moroz, Y. S., Korendovych, I. V., and Yin, H. (2012) Isolated Toll-like receptor transmembrane domains are capable of oligomerization. PLoS One 7, e48875. [30] Fink, A., Reuven, E. M., Arnusch, C. J., Shmuel-Galia, L., Antonovsky, N., and Shai, Y. (2013) Assembly of the TLR2/6 transmembrane domains is essential for activation and is a target for prevention of sepsis. J. Immunol. 190, 6410-6422. [31] Shmuel-Galia, L., Aychek, T., Fink, A., Porat, Z., Zarmi, B., Bernshtein, B., et al. (2016) Neutralization of pro-inflammatory monocytes by targeting TLR2 dimerization ameliorates colitis. EMBO J. 35, 685-698. [32] Lee, J., Miyazaki, M., Romeo, G. R., and Shoelson, S. E. (2014) Insulin receptor activation with transmembrane domain ligands. J. Biol. Chem. 289, 19769-19777. [33] Taniguchi, C. M., Emanuelli, B., and Kahn, C. R. (2006) Critical nodes in signalling pathways: insights into insulin action. Nat. Rev. Mol. Cell Biol. 7, 85-96. [34] Lemmon, M. A., and Schlessinger, J. (2010) Cell signaling by receptor tyrosine kinases. Cell 141, 11171134. [35] Cohen, D. H., and LeRoith, D. (2012) Obesity, type 2 diabetes, and cancer: the insulin and IGF connection. Endocr.-Relat. Cancer 19, F27-F45. [36] Lemmon, M. A., Flanagan, J. M., Treutlein, H. R., Zhang, J., and Engelman, D. M. (1992) Sequence specificity in the dimerization of transmembrane alpha-helices. Biochemistry 31, 12719-12725.

ACS Paragon Plus Environment

Biochemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 28 of 42

[37] Treutlein, H. R., Lemmon, M. A., Engelman, D. M., and Brunger, A. T. (1992) The glycophorin-A transmembrane domain dimer- sequence-specific propensity for a right-handed supercoil of helices. Biochemistry 31, 12726-12733. [38] Arkhipov, A., Shan, Y. B., Das, R., Endres, N. F., Eastwood, M. P., Wemmer, D. E., et al. (2013) Architecture and membrane interactions of the EGF receptor. Cell 152, 557-569. [39] Mineev, K. S., Bocharov, E. V., Pustovalova, Y. E., Bocharova, O. V., Chupin, V. V., and Arseniev, A. S. (2010) Spatial structure of the transmembrane domain heterodimer of ErbB1 and ErbB2 receptor tyrosine kinases. J. Mol. Biol. 400, 231-243. [40] Tao, R. H., and Maruyama, I. N. (2008) All EGF(ErbB) receptors have preformed homo- and heterodimeric structures in living cells. J. Cell Sci. 121, 3207-3217. [41] Riese, D. J., Gallo, R. M., and Settleman, J. (2007) Mutational activation of ErbB family receptor tyrosine kinases: insights into mechanisms of signal transduction and tumorigenesis. Bioessays 29, 558-565. [42] Citri, A., and Yarden, Y. (2006) EGF-ERBB signalling: towards the systems level. Nat. Rev. Mol. Cell Biol. 7, 505-516. [43] Schlessinger, J. (2002) Ligand-induced, receptor-mediated dimerization and activation of EGF receptor. Cell 110, 669-672. [44] Lemmon, M. A., Bu, Z. M., Ladbury, J. E., Zhou, M., Pinchasi, D., Lax, I., et al. (1997) Two EGF molecules contribute additively to stabilization of the EGFR dimer. EMBO J. 16, 281-294. [45] Gerber, D., Sal-Man, N., and Shai, Y. (2004) Two motifs within a transmembrane domain, one for homodimerization and the other for heterodimerization. J. Biol. Chem. 279, 21177-21182. [46] Bennasroune, A., Fickova, M., Gardin, A., Dirrig-Grosch, S., Aunis, D., Cremel, G., et al. (2004) Transmembrane peptides as inhibitors of ErbB receptor signaling. Mol. Biol. Cell 15, 3464-3474. [47] Jaroniec, C. P., Kaufman, J. D., Stahl, S. J., Viard, M., Blumenthal, R., Wingfield, P. T., et al. (2005) Structure and dynamics of micelle-associated human immunodeficiency virus gp41 fusion domain. Biochemistry 44, 16167-16180.

ACS Paragon Plus Environment

Page 29 of 42

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biochemistry

[48] Kliger, Y., Aharoni, A., Rapaport, D., Jones, P., Blumenthal, R., and Shai, Y. (1997) Fusion peptides derived from the HIV type 1 glycoprotein 41 associate within phospholipid membranes and inhibit cell-cell fusion - Structure-function study. J. Biol. Chem. 272, 13496-13505. [49] Pritsker, M., Jones, P., Blumenthal, R., and Shai, Y. C. (1998) A synthetic all D-amino acid peptide corresponding to the N-terminal sequence of HIV-1 gp41 recognizes the wild-type fusion peptide in the membrane and inhibits HIV-1 envelope glycoprotein-mediated cell fusion. Proc. Natl. Acad. Sci. U. S. A. 95, 7287-7292. [50] Gerber, D., and Shai, Y. (2002) Chirality-independent protein-protein recognition between transmembrane domains in vivo. J. Mol. Biol. 322, 491-495. [51] Gerber, D., Sal-Man, N., and Shai, Y. (2004) Structural adaptation of the glycophorin a transmembrane homodimer to D-amino acid modifications. J. Mol. Biol. 339, 243-250. [52] Manolios, N., Collier, S., Taylor, J., Pollard, J., Harrison, L. C., and Bender, V. (1997) T-cell antigen receptor transmembrane peptides modulate T-cell function and T cell-mediated disease. Nat. Med. 3, 84-88. [53] Gerber, D., Quintana, F. J., Bloch, I., Cohen, I. R., and Shai, Y. (2005) D-enantiomer peptide of the TCR alpha transmembrane domain inhibits T-cell activation in vitro and in vivo. FASEB J. 19, 1190-1192. [54] Quintana, F. J., Gerber, D., Bloch, I., Cohen, I. R., and Shai, Y. (2007) A structurally altered D,L-amino acid TCR alpha transmembrane peptide interacts with the TCR alpha and inhibits T-cell activation in vitro and in an animal model. Biochemistry 46, 2317-2325. [55] Yin, H., Slusky, J. S., Berger, B. W., Walters, R. S., Vilaire, G., Litvinov, R. I., et al. (2007) Computational design of peptides that target transmembrane helices. Science 315, 1817-1822. [56] Senes, A., Chadi, D. C., Law, P. B., Walters, R. F. S., Nanda, V., and DeGrado, W. F. (2007) E-z, a depthdependent potential for assessing the energies of insertion of amino acid side-chains into membranes: Derivation and applications to determining the orientation of transmembrane and interfacial helices. J. Mol. Biol. 366, 436448. [57] Caputo, G. A., Litvinov, R. I., Li, W., Bennett, J. S., DeGrado, W. F., and Yin, H. (2008) Computationally designed peptide inhibitors of protein-protein interactions in membranes. Biochemistry 47, 8600-8606.

ACS Paragon Plus Environment

Biochemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 30 of 42

[58] Kritzer, J. A., Stephens, O. M., Guarracino, D. A., Reznik, S. K., and Schepartz, A. (2005) beta-Peptides as inhibitors of protein-protein interactions. Bioorg. Med. Chem. 13, 11-16. [59] Shandler, S. J., Korendovych, I. V., Moore, D. T., Smith-Dupont, K. B., Streu, C. N., Litvinov, R. I., et al. (2011) Computational Design of a beta-Peptide That Targets Transmembrane Helices. J. Am. Chem. Soc. 133, 12378-12381. [60] Burkhardt, A., Dimaio, D., and Schlegel, R. (1987) Genetic and biochemical definition of the bovine papillomavirus E5 transforming protein. EMBO J. 6, 2381-2385. [61] Talbert-Slagle, K., and DiMaio, D. (2009) The bovine papillomavirus E5 protein and the PDGF beta receptor: It takes two to tango. Virology 384, 345-351. [62] Cammett, T. J., Jun, S. J., Cohen, E. B., Barrera, F. N., Engelman, D. M., and DiMaio, D. (2010) Construction and genetic selection of small transmembrane proteins that activate the human erythropoietin receptor. Proc. Natl. Acad. Sci. U. S. A. 107, 3447-3452. [63] Scheideman, E. H., Marlatt, S. A., Xie, Y. H., Hu, Y. N., Sutton, R. E., and DiMaio, D. (2012) Transmembrane protein aptamers that inhibit CCR5 expression and HIV coreceptor function. J. Virol. 86, 10281-10292. [64] Millan, M. J. (2002) Descending control of pain. Prog. Neurobiol. 66, 355-474. [65] Bohn, L. M., Gainetdinov, R. R., Lin, F. T., Lefkowitz, R. J., and Caron, M. G. (2000) mu-Opioid receptor desensitization by beta-arrestin-2 determines morphine tolerance but not dependence. Nature 408, 720-723. [66] Bohn, L. M., Lefkowitz, R. J., Gainetdinov, R. R., Peppel, K., Caron, M. G., and Lin, F. T. (1999) Enhanced morphine analgesia in mice lacking beta-arrestin 2. Science 286, 2495-2498. [67] Raehal, K. M., Walker, J. K. L., and Bohn, L. M. (2005) Morphine side effects in beta-arrestin 2 knockout mice. J. Pharmacol. Exp. Ther. 314, 1195-1201. [68] Manglik, A., Lin, H., Aryal, D. K., McCorvy, J. D., Dengler, D., Corder, G., et al. (2016) Structure-based discovery of opioid analgesics with reduced side effects. Nature 537, 185-190. [69] Roth, B. L., and Kroeze, W. K. (2015) Integrated Approaches for genome-wide interrogation of the druggable non-olfactory G Protein-coupled receptor superfamily. J. Biol. Chem. 290, 19471-19477.

ACS Paragon Plus Environment

Page 31 of 42

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biochemistry

[70] Huang, X. P., Karpiak, J., Kroeze, W. K., Zhu, H., Chen, X., Moy, S. S., et al. (2015) Allosteric ligands for the pharmacologically dark receptors GPR68 and GPR65. Nature 527, 477-483. [71] Deng, D., Xu, C., Sun, P. C., Wu, J. P., Yan, C. Y., Hu, M. X., et al. (2014) Crystal structure of the human glucose transporter GLUT1. Nature 510, 121-126. [72] Deng, D., Sun, P. C., Yan, C. Y., Ke, M., Jiang, X., Xiong, L., et al. (2015) Molecular basis of ligand recognition and transport by glucose transporters. Nature 526, 391-398. [73] Nomura, N., Verdon, G., Kang, H. J., Shimamura, T., Nomura, Y., Sonoda, Y., et al. (2015) Structure and mechanism of the mammalian fructose transporter GLUT5. Nature 526, 397-401. [74] George Thompson, A. M., Ursu, O., Babkin, P., Iancu, C. V., Whang, A., Oprea, T. I., et al. (2016) Discovery of a specific inhibitor of human GLUT5 by virtual screening and in vitro transport evaluation. Sci. Rep. 6, 24240. [75] Mishra, R. K., Wei, C. Y., Hresko, R. C., Bajpai, R., Heitmeier, M., Matulis, S. M., et al. (2015) In silico modeling-based identification of glucose transporter 4 (GLUT4)-selective inhibitors for cancer therapy. J. Biol. Chem. 290, 14441-14453. [76] Hurdle, J. G., O'Neill, A. J., Chopra, I., and Lee, R. E. (2011) Targeting bacterial membrane function: an underexploited mechanism for treating persistent infections. Nat. Rev. Microbiol. 9, 62-75. [77] Andries, K., Verhasselt, P., Guillemont, J., Gohlmann, H. W. H., Neefs, J. M., Winkler, H., et al. (2005) A diarylquinoline drug active on the ATP synthase of Mycobacterium tuberculosis. Science 307, 223-227. [78] Biukovic, G., Basak, S., Manimekalai, M. S. S., Rishikesan, S., Roessle, M., Dick, T., et al. (2013) Variations of Subunit epsilon of the Mycobacterium tuberculosis F1Fo ATP Synthase and a Novel Model for Mechanism of Action of the Tuberculosis Drug TMC207. Antimicrob. Agents Chemother. 57, 168-176. [79] Times, N. Y. (2012) F.D.A. Approves drug for resistant tuberculosis, (Thomas, K., Ed.). [80] Toussirot, E., and Roudier, J. (2008) Epstein-Barr virus in autoimmune diseases. Best Pract. Res. Clin. Rheumatol. 22, 883-896. [81] Kuppers, R. (2005) Mechanisms of B-cell lymphoma pathogenesis. Nat. Rev. Cancer 5, 251-262.

ACS Paragon Plus Environment

Biochemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 32 of 42

[82] Sammond, D. W., Joce, C., Takeshita, R., McQuate, S. E., Ghosh, N., Martin, J. M., et al. (2011) Transmembrane peptides used to investigate the homo-oligomeric interface and binding hotspot of latent membrane protein 1. Biopolymers 95, 772-784. [83] Wang, X. H., Saludes, J. P., Zhao, T. X., Csakai, A., Fiorini, Z., Chavez, S. A., et al. (2012) Targeting the lateral interactions of transmembrane domain 5 of Epstein-Barr virus latent membrane protein 1. Biochim. Biophys. Acta-Biomembr. 1818, 2282-2289.

ACS Paragon Plus Environment

Page 33 of 42

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biochemistry

For Table of Contents Use Only Title: Small molecule and peptide recognition of protein transmembrane domains Authors: Xianfeng Zeng, Peiyao Wu, Chengbo Yao, Jiaqi Liang, Shuting Zhang, Hang Yin

ACS Paragon Plus Environment

Biochemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Overviews of major methods to identify agents recognizing MP TMDs. 174x109mm (144 x 144 DPI)

ACS Paragon Plus Environment

Page 34 of 42

Page 35 of 42

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biochemistry

Rational design approaches based on natural product scaffolds. One of the approaches for the discovery of new agents recognizing protein TMDs is to attach a new pharmacophore to natural small molecules (left). The high affinity between nature-existed molecules and their TMD targets would guide the new attached pharmacophore to the binding sites; Also, structural modifications on natural small molecules could improve the affinity between those natural product analogs and TMD target (right). 288x116mm (144 x 144 DPI)

ACS Paragon Plus Environment

Biochemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

The structure of selective TGR5 agonists 1-3. Compounds 1-3 were obtained by replacing hydroxyl group with amino group in scaffold of LCA. 154x77mm (144 x 144 DPI)

ACS Paragon Plus Environment

Page 36 of 42

Page 37 of 42

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biochemistry

Warburg effect and strategies of developing small-molecule drugs exploiting Warburg effect. (A) Warburg effect: GLUTs are overexpressed in cancer cells to compensate for low metabolic efficiency, and enable unregulated cell growth; (B) Strategies exploiting Warburg effect: Glucose conjugates enable selective delivery of anticancer pharmacophores to the cancer cells (left); GLUTs inhibitors can directly block the glucose uptake in energy demanding cancer cells (right); (C) The structure of designed hexose conjugates and their aglycone moiety. 167x245mm (144 x 144 DPI)

ACS Paragon Plus Environment

Biochemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

The molecular structure of globomycin and its co-crystal structure of globomycin in complex with LspA. (A) The structure of globomycin (8) and its analog 9; (B) The crystal structure of LspA-globomycin complex. (PDB entry: 5DIR). Modified from Reference 20. Figure 5B is reprinted with permission. 313x116mm (144 x 144 DPI)

ACS Paragon Plus Environment

Page 38 of 42

Page 39 of 42

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biochemistry

TOXCAT system and truncated peptides as probes recognizing MP TMDs. (A) TOXCAT system: MBP: maltose binding protein; TM1 and TM2: Two engineered transmembrane helices; TM1 and TM2 interact with each other and form a functional ToxR dimer. The ToxR dimer binds to ctx promoter and induces measurable synthesis of CAT. TOXCAT provides a general way to determine the heterodimerization of two transmembrane helices; (B) Truncated peptides: Once the interacting protein transmembrane helices are determined, the truncated TMDs of one oligo partner can be exploited to modulate native oligomerization. 209x176mm (144 x 144 DPI)

ACS Paragon Plus Environment

Biochemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Targeting the TMDs with optical isomers. D-enantiomers possess same amino acid sequence but different residue orientations comparing with their L-counterpart, by changing the tilt angle between two helices, it is possible to bring the similar interacting surfaces orientation as the L-homodimer did and with some special stability and affinity to their TMD target. 268x183mm (144 x 144 DPI)

ACS Paragon Plus Environment

Page 40 of 42

Page 41 of 42

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biochemistry

Design of CHAMP peptides targeting TMDs. (A) The general procedure to predict CHAMP peptide interface: The first step is to select a backbone geometry from the known structures that contain motifs in TMD target. Then amino acid residues are added to backbone, followed by a side chain-repacking algorithm; (B) The sequence motifs of αIIbβ3 and αVβ3; (C) The predicted packing interface between the integrin and CHAMP; (D) Activation of integrins by CHAMP peptide. Modified from Reference 55. Figure 8 is reprinted with permission. 181x89mm (144 x 144 DPI)

ACS Paragon Plus Environment

Biochemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Targeting the TMDs by traptmers. Traptmer library can be constructed by replacing the TMD of E5 peptide with hydrophobic residues. High affinity traptmers for the target MP TMD will be identified by genetic screening. 120x130mm (220 x 220 DPI)

ACS Paragon Plus Environment

Page 42 of 42