throbber
Structure, Volume 20
`
`Supplemental Information
`
`Staphylococcus aureus FabI: Inhibition, Substrate
`Recognition, and Potential Implications
`for In Vivo Essentiality
`Johannes Schiebel, Andrew Chang, Hao Lu, Michael V. Baxter, Peter J. Tonge,
`Caroline Kisker
`
`Inventory of Supplemental Information
`Inventory of Supplemental Information ................................................................................ 1 
`Supplemental Data ................................................................................................................... 2 
`Figure S1, related to Figure 1. Substrate and inhibitor structures. ............................ 2 
`Figure S2, related to Figure 2. Inhibitor and cofactor electron densities. .................. 3 
`Figure S3, related to Figure 6. Quaternary structure of saFabI in solution. .............. 4 
`Figure S4, related to Figure 5. Active site formation. ................................................... 5 
`Figure S5, related to Figure 6. Cooperativity of substrate binding to saFabI. ........... 6 
`Table S1, related to Experimental Procedures. ............................................................. 8 
`Table S2, related to Experimental Procedures. ............................................................. 9 
`Table S3, related to Table 3. .......................................................................................... 10 
`Table S4, related to Experimental Procedures. ........................................................... 11 
`Movie S1, related to Figure 5. Ordering of the substrate binding loop region. ........ 13 
`Movie S2, related to Figure 6. Dimer - tetramer transition. ...................................... 13 
`Supplemental Results ............................................................................................................. 14 
`Inhibition kinetics ............................................................................................................... 14 
`Reduction of straight-chain substrates by saFabI ........................................................... 14 
`Substrate specificity of saFabI .......................................................................................... 15 
`Supplemental Discussion ....................................................................................................... 16 
`Glutamate sensitivity .......................................................................................................... 16 
`Supplemental Experimental Procedures .............................................................................. 18 
`Cloning ................................................................................................................................ 18 
`Expression and purification .............................................................................................. 18 
`Crystallization, data collection and structure determination ........................................ 19 
`Steady-state kinetic assays ................................................................................................. 21 
`Inhibition kinetics ............................................................................................................... 23 
`Supplemental References ....................................................................................................... 24 
`
`1
`
`Chugai Exhibit 2305
`Pfizer, Inc. v. Chugai Pharmaceutical Co. Ltd.
`IPR2017-01357
`Page 00001
`
`

`

`Supplemental Data
`
`
`
`
`
`Figure S1, related to Figure 1. Substrate and inhibitor structures. The chemical structures
`
`of the saFabI substrates and inhibitors, which were analyzed in this study, are shown.
`
`2
`
`Chugai Exhibit 2305
`Pfizer, Inc. v. Chugai Pharmaceutical Co. Ltd.
`IPR2017-01357
`Page 00002
`
`

`

`
`
`
`
`Figure S2, related to Figure 2. Inhibitor and cofactor electron densities. (A-C) 2Fo-Fc
`
`omit maps of triclosan (CPP, EPP) and NADP+ in the TCL-2 (CPP, EPP) structure (subunit
`
`H) contoured at one time the standard deviation of the mean electron density.
`
`3
`
`Chugai Exhibit 2305
`Pfizer, Inc. v. Chugai Pharmaceutical Co. Ltd.
`IPR2017-01357
`Page 00003
`
`

`

`
`
`
`
`Figure S3, related to Figure 6. Quaternary structure of saFabI in solution. (A) Dimeric
`
`and tetrameric structure of saFabI. Overlay of two different analytical SEC chromatograms
`
`using buffers at pH 5.6 (20 mM trisodium citrate pH 5.6, 500 mM NaCl and 100 mM EDTA)
`
`shown in red, or at pH 8.0 (20 mM Tris pH 8.0 and 200 mM NaCl) shown in blue,
`
`respectively. (B) Dimer - tetramer transition. Four different analytical SEC chromatograms
`
`and the respective cropped SDS-PAGE gel bands for the peak fractions (elution volumes of
`
`14.0 and 16.0 ml) running at the same height (~33 kDa, calculated monomeric saFabI
`
`molecular mass is 31 kDa) are shown with apo saFabI in black, and saFabI treated with
`
`NADP+, NADPH or NADP+ and TCL in red, orange or blue, respectively. The front peak
`
`corresponds to a molecular mass of 131 kDa, the middle peak to 47 kDa. Excess of unbound
`
`NADP+ and NADPH elutes at 19.8 ml.
`
`4
`
`Chugai Exhibit 2305
`Pfizer, Inc. v. Chugai Pharmaceutical Co. Ltd.
`IPR2017-01357
`Page 00004
`
`

`

`
`
`
`
`Figure S4, related to Figure 5. Active site formation. Superposition of the apo-2 (blue,
`
`subunit A) and TCL-2 (yellow) structures. All three active site residues are depicted as stick
`
`representations for both structures. TCL and NADP+ are shown in grey. The three active site
`
`residues (Tyr147, Tyr157, Lys164) move significantly upon cofactor and inhibitor binding.
`
`Tyr147 is shifted by ~5 Å out of the nicotinamide binding pocket whereas Tyr157 rotates
`
`about 130° towards the TCL hydroxyl group (Priyadarshi et al., 2010; Rozwarski et al., 1999).
`
`At the same time, the 80° rotation of Lys164 allows the correct placement of the cofactors
`
`nicotinamide ring (Rozwarski et al., 1999).
`
`5
`
`Chugai Exhibit 2305
`Pfizer, Inc. v. Chugai Pharmaceutical Co. Ltd.
`IPR2017-01357
`Page 00005
`
`

`

`
`
`
`
`Figure S5, related to Figure 6. Cooperativity of substrate binding to saFabI. Reaction
`
`velocities with 22 nM saFabI are plotted as a function of substrate concentration, holding the
`
`other substrate concentration constant. (A) In the absence of potassium glutamate, cooperative
`
`binding of NADPH at 100 µM trans-2-octenoyl-CoA can be observed. To approximate the
`
`Hill coefficient (h) despite the truncated data set, best-fit curves to h = 1, 2, 3 and 4 are
`
`displayed. Note that h = 2.2 was previously reported for the binding of NADPH to saFabI
`
`(Heath et al., 2000). (B) In the presence of 1 M potassium glutamate, h = 0.9  0.1 for the
`
`binding of NADPH at 100 µM trans-2-octenoyl-CoA. (C) In the absence of potassium
`
`glutamate, h = 2.3  0.3 for the binding of trans-2-octenoyl-CoA at 350 µM NADPH (R2 =
`
`6
`
`Chugai Exhibit 2305
`Pfizer, Inc. v. Chugai Pharmaceutical Co. Ltd.
`IPR2017-01357
`Page 00006
`
`

`

`0.99). (D) In the presence of 250 mM, 500 mM and 1 M potassium glutamate, h = 2.4  0.4,
`
`1.4  0.3 and 1.1  0.3, respectively, for the binding of trans-2-octenoyl-CoA at 350 µM
`
`NADPH (R2 = 0.99 for each curve-fit). In addition, note the rise in Vmax as the concentration
`
`of glutamate increases. (E) Potential glutamate binding site. The ambiguous density of the
`
`2Fo-Fc omit map (CPP structure, subunit G) contoured at one time the standard deviation of
`
`the mean electron density is shown between Arg103 located on SBL-2 (cyan) and Asn205
`
`located on SBL (red). The remainder of the protein is omitted for clarity.
`
`7
`
`Chugai Exhibit 2305
`Pfizer, Inc. v. Chugai Pharmaceutical Co. Ltd.
`IPR2017-01357
`Page 00007
`
`

`

`Table S1, related to Experimental Procedures. Data collection and refinement statistics
`
`(apo structures).
`
`
`Data collection
`Cell dimensions
`a, b, c (Å)
`α, β, γ (°)
`Space group
`Resolution1 (Å)
`Observed reflections
`Unique reflections
`Completeness (%)
`Average redundancy
`2 (%)
`Rmerge
`<I / σ(I)>
`Monomers per AU
`Refinement
`Resolution (Å)
`3
` (%)
`Rcryst
`Rfree (%)
`Twin fraction
`Number of atoms
`rmsd bond lengths (Å)
`rmsd bond angles (°)
`Average B-factor (Å2)
`Ramachandran-plot4
`Favored (%)
`Allowed (%)
`Outliers (%)
`Maximum likelihood based
`estimated coordinate error (Å)
`PDB code
`
`Apo-1
`
`
`123.8, 123.8, 190.1
`90.0, 90.0, 120.0
`P32
`50.0-3.05 (3.20-3.05)
`160,521 (21,802)
`61,623 (8,296)
`99.3 (99.7)
`2.6 (2.6)
`11.7 (49.9)
`6.8 (2.1)
`12
`
`44.3-3.05
`20.3
`24.6
`0.33
`20,062
`0.015
`1.482
`67.1
`
`91.5
`7.9
`0.6
`0.28
`
`Apo-2
`
`
`87.5, 87.5, 307.2
`90.0, 90.0, 90.0
`P43212
`50.0-2.45 (2.60-2.45)
`319,536 (52,121)
`44,583 (7,185)
`98.9 (99.8)
`7.2 (7.3)
`8.8 (78.0)
`16.4 (2.8)
`4
`
`48.2-2.45
`18.5
`25.1
`-
`7,820
`0.007
`0.980
`62.9
`
`96.7
`3.1
`0.2
`0.30
`
`4ALN
`
`4ALM
`
`
`
`1Values in parenthesis refer to the highest resolution shell
`
`2
`
`R
`merge
`
`
`
` 
`I
`
`hkl
`
`i
`
`i
`
`I
`
`
`
`hkl
`
`i
`
`I
`
`i
`
`
`
`3
`
`R
`cryst
`
`
`
` 
`F
`obs
`
`hkl
`
`F
`calc
`
`F
`obs
`
`
`
`
`
`hkl
`
`4According to Molprobity (Davis et al., 2007)
`
`
`
`8
`
`Chugai Exhibit 2305
`Pfizer, Inc. v. Chugai Pharmaceutical Co. Ltd.
`IPR2017-01357
`Page 00008
`
`

`

`Table S2, related to Experimental Procedures. Data collection and refinement statistics
`
`(ternary complex structures).
`
`
`Data collection
`Cell dimensions
`a, b, c (Å)
`α, β, γ (°)
`Space group
`Resolution1 (Å)
`Observed reflections
`Unique reflections
`Completeness (%)
`Average redundancy
`2 (%)
`Rmerge
`3 (%)
`Rpim
`<I / σ(I)>
`Monomers per AU
`Refinement
`Resolution (Å)
`4
` (%)
`Rcryst
`Rfree (%)
`Twin fraction
`Number of atoms
`rmsd bond lengths (Å)
`rmsd bond angles (°)
`Average B-factor (Å2)
`Ramachandran-plot5
`Favored (%)
`Allowed (%)
`Outliers (%)
`Maximum likelihood based
`estimated coordinate error (Å)
`PDB code
`
`
`
`TCL-1
`
`
`83.5, 111.9, 111.6
`90.0, 90.0, 90.0
`P212121
`42.9-2.80 (2.95-2.80)
`119,875 (17,343)
`22,827 (3,361)
`87.2 (89.6)
`5.3 (5.2)
`15.3 (35.7)
`6.8 (16.2)
`6.5 (2.5)
`4
`
`79.0-2.80
`21.4
`26.2
`-
`8,101
`0.013
`1.591
`66.4
`
`96.2
`3.6
`0.2
`0.38
`
`TCL-2
`
`
`90.0, 94.8, 94.9
`98.1, 112.0, 97.3
`P1
`38.5-2.10 (2.21-2.10)
`640,244 (92,983)
`161,229 (23,385)
`97.8 (97.0)
`4.0 (4.0)
`14.1 (84.3)
`8.2 (48.8)
`8.0 (2.0)
`8
`
`38.5-2.10
`14.9
`19.8
`-
`18,131
`0.013
`1.839
`31.3
`
`96.3
`3.6
`0.1
`0.12
`
`CPP
`
`
`90.2, 94.8, 94.8
`98.0, 112.4, 97.4
`P1
`49.1-2.20 (2.32-2.20)
`249,699 (35,984)
`135,192 (19,491)
`94.4 (93.2)
`1.8 (1.8)
`8.2 (34.8)
`8.2 (34.8)
`5.9 (1.9)
`8
`
`49.1-2.20
`15.8
`21.9
`-
`17,947
`0.012
`1.788
`47.7
`
`96.8
`3.2
`0.0
`0.14
`
`EPP
`
`
`90.2, 95.1, 95.3
`98.2, 112.3, 97.3
`P1
`49.3-1.90 (2.00-1.90)
`427,322 (63,042)
`206,602 (30,427)
`92.1 (92.6)
`2.1 (2.1)
`6.9 (40.4)
`6.4 (38.0)
`6.4 (2.0)
`8
`
`40.9-1.90
`15.0
`19.3
`-
`18,573
`0.015
`1.767
`30.1
`
`97.0
`3.0
`0.0
`0.09
`
`4ALL
`
`4ALI
`
`4ALJ
`
`4ALK
`
`1Values in parenthesis refer to the highest resolution shell
`
`2
`
`R
`merge
`
`
`
` 
`I
`
`hkl
`
`i
`
`i
`
`I
`
`
`
`hkl
`
`i
`
`3
`
`R
`
`pim
`
`
`
`
`
`hkl
`
`
`1
`
`
`
`N
`
`
`
`2/11
`
`
`
` 
`I
`
`i
`
`i
`
`I
`
`i
`
`
`
`I
`
`
`
`hkl
`
`i
`
`I
`
`i
`
` (Weiss, 2001)
`
`4
`
`R
`cryst
`
`
`
` 
`F
`obs
`
`hkl
`
`F
`calc
`
`F
`obs
`
`
`
`
`
`hkl
`
`5According to Molprobity (Davis et al., 2007)
`
`
`
`9
`
`Chugai Exhibit 2305
`Pfizer, Inc. v. Chugai Pharmaceutical Co. Ltd.
`IPR2017-01357
`Page 00009
`
`

`

`Table S3, related to Table 3. Kinetic parameters for ftFabI utilizing NADH and various
`
`trans-2-enoyl-CoA substrates.
`
`
`
`trans-2-butenoyl-CoA
`
`Km (µM)
`
`353  5
`
`kcat (min-1)
`
`kcat/Km,acyl-CoA (min-1 µM-1)
`
`1140  60
`
`3.2  0.2
`
`trans-2-octenoyl-CoA
`
`33.3  0.4
`
`1200  60
`
`36.0  1.9
`
`trans-2-decenoyl-CoA
`
`11.2  0.8
`
`1260  60
`
`112.5  9.7
`
`trans-2-dodecenoyl-CoA
`
`1.7  0.1
`
`1200  60
`
`705.9  54.5
`
`trans-5-methyl-2-
`
`hexenoyl-CoA (iso-)
`
`()-trans-4-methyl-2-
`
`
`
`
`
`hexenoyl-CoA (anteiso-)
`
`
`
`
`
`0.287  0.0071
`
`0.0040  0.00021
`
`NADH
`
`
`
`18.8  0.6
`
`1140  60
`
`60.0  6.0
`
`1Estimated based on linear slope of the Michaelis-Menten plot at low [NADH]
`
`
`
`10
`
`Chugai Exhibit 2305
`Pfizer, Inc. v. Chugai Pharmaceutical Co. Ltd.
`IPR2017-01357
`Page 00010
`
`

`

`Table S4, related to Experimental Procedures. Details of the saFabI purification protocols.*
`
`A
`pET-16b(+)
`BL21 (DE3)
`pLysS
`100 µg/ml
`ampicillin and 34
`µg/ml
`chloramphenicol
`28 °C, 5 h
`
`B
`pETM-11
`BL21 (DE3)
`
`50 µg/ml
`kanamycin
`
`C
`pETM-112
`BL21 (DE3)
`
`50 µg/ml
`kanamycin2
`
`D
`pETM-11
`BL21 (DE3)
`
`50 µg/ml
`kanamycin
`
`E
`pETM-11
`BL21 (DE3)
`
`50 µg/ml
`kanamycin
`
`18 °C, 18 h
`
`18 °C, 18 h2
`
`18 °C, 18 h
`
`18 °C, 18 h
`
`
`Plasmid
`E. coli strain
`
`antibiotics
`
`cultivation
`temperature,
`time
`lysis buffer
`
`wash buffer
`
`elution buffer
`
`50 mM Tris-HCl
`pH 8.0, 500 mM
`NaCl
`
`50 mM Tris-HCl
`pH 8.0, 500 mM
`NaCl
`
`50 mM Tris-HCl
`pH 8.0, 500 mM
`NaCl
`
`50 mM Tris-HCl
`pH 8.0, 500 mM
`NaCl
`
`50 mM Tris-HCl
`pH 8.0, 500 mM
`NaCl, 10 mM
`imidazole
`
`100 mM
`trisodium citrate
`pH 5.6, 500 mM
`NaCl, 100 mM
`EDTA
`
`50 mM Tris-HCl
`pH 8.0, 1 M NaCl
`
`50 mM Tris-HCl
`pH 8.0, 1 M
`NaCl2
`
`50 mM Tris-HCl
`pH 8.0, 1 M NaCl
`
`50 mM Tris-HCl
`pH 8.0, 500 mM
`NaCl, 250 mM
`imidazole
`
`50 mM Tris-HCl
`pH 8.0, 500 mM
`NaCl, 250 mM
`imidazole
`
`50 mM Tris-HCl
`pH 8.0, 500 mM
`NaCl, 250 mM
`imidazole
`
`50 mM Tris-HCl
`pH 8.0, 200 mM
`NaCl, 2 mM β-
`mercaptoethanol,
`5% glycerol
`50 mM Tris-HCl
`pH 8.0, 500 mM
`NaCl, 2 mM β-
`mercaptoethanol,
`5% glycerol
`50 mM Tris-HCl
`pH 8.0, 200 mM
`NaCl, 2 mM β-
`mercaptoethanol,
`5% glycerol, 250
`mM imidazole
`no
`25 mM Tris-HCl
`pH 8.0, 200 mM
`NaCl, 1 mM DTT
`
`15.2 mg/ml (0.49
`mM)
`
`TEV cleavage
`storage buffer
`
`no
`20 mM trisodium
`citrate pH 5.6,
`500 mM NaCl,
`100 mM EDTA
`
`yes1
`20 mM trisodium
`citrate pH 5.6,
`500 mM NaCl,
`100 mM EDTA
`
`no
`20 mM trisodium
`citrate pH 5.6,
`500 mM NaCl,
`100 mM EDTA3
`
`9.8 mg/ml (0.32
`mM)
`
`38.6 mg/ml (1.24
`mM)
`
`13.0 mg/ml (0.42
`mM) 2,3
`
`no
`20 mM trisodium
`citrate pH 5.6,
`280 mM NaCl, 1
`mM EDTA, 280
`mM K-glutamate
`15.0 mg/ml (0.48
`mM)
`
`final
`concentration
`
`1After Ni2+ affinity chromatography the protein sample was incubated with TEV-protease in
`
`50 mM Tris pH 8.0 and 500 mM NaCl at 4 °C for 10 h, yielding a 50% removal of the His6-
`
`tag. Subsequently, the sample was re-applied to Ni-TED followed by size exclusion
`
`chromatography as described above.
`
`
`
`2Alternatively (C'), the saFabI mutants were expressed and purified (final concentrations of
`
`1.0 mg/ml) using the pET-16b(+) plasmid, 100 µg/ml ampicillin, a cultivation temperature of
`
`25 °C and the wash buffer described in procedure A.
`
`
`
`11
`
`Chugai Exhibit 2305
`Pfizer, Inc. v. Chugai Pharmaceutical Co. Ltd.
`IPR2017-01357
`Page 00011
`
`

`

`3Alternatively (C''), the storage buffer of the pooled SEC-fractions was exchanged to 20 mM
`
`trisodium citrate pH 5.6, 150 mM NaCl and 1 mM EDTA before concentrating to 8.5 mg/ml
`
`(0.27 mM).
`
`
`
`*In general, the procedure described in the Supplemental Experimental Procedures was
`
`applied to obtain ≥ 95% pure saFabI protein. The crystals leading to the TCL-1, apo-1 and
`
`apo-2 structures were obtained using purifications A, B or C'', respectively. For the EPP,
`
`TCL-2 and CPP complex structures a FabI batch was used which was obtained by procedure
`
`D. The analytical size exclusion chromatography experiments were performed with protein
`
`batches E and C. The double and triple mutants were purified applying procedure C'.
`
`12
`
`Chugai Exhibit 2305
`Pfizer, Inc. v. Chugai Pharmaceutical Co. Ltd.
`IPR2017-01357
`Page 00012
`
`

`

`Movie S1, related to Figure 5. Ordering of the substrate binding loop region. This model
`
`is based on the TCL-2 structure (one monomer is shown in cyan, TCL in yellow and NADP+
`
`in green). Helix α7 was rearranged in PyMol according to the different geometries of the apo-
`
`2 structure. The motion of helix α7 was visualized by interpolating between these different
`
`arrangements. The loop region between α7 and α8 was omitted for clarity (the associated
`
`termini are shown in red). The initial states of the SBL (shown in green), cofactor and
`
`inhibitor were not obtained directly in our apo structures. Conformational changes in
`
`additional regions (SBL-2 and ASL) were omitted in the animation. This animation was
`
`prepared using PyMol, VirtualDub and Xvid (DeLano, 2002).
`
`
`
`Movie S2, related to Figure 6. Dimer - tetramer transition. This animation was prepared
`
`using the structures 3GNT, 3GNS, apo-1, apo-2 and TCL-2. One dimer is shown in blue, the
`
`other in red. All structures were superimposed using the lower left monomer. LSQMAN was
`
`used to interpolate between Cα positions of consecutive structure pairs for the amino acids
`
`which were ordered in both structures (Kleywegt and Jones, 1996). Cα traces of these
`
`consecutive states were visualized using PyMol, VirtualDub and Xvid (DeLano, 2002).
`
`13
`
`Chugai Exhibit 2305
`Pfizer, Inc. v. Chugai Pharmaceutical Co. Ltd.
`IPR2017-01357
`Page 00013
`
`

`

`Supplemental Results
`
`Inhibition kinetics
`
`Based on the relative Ki*,app measurements, the gain in affinity from the additional buried
`
`surface area (BSA) for the inhibitors CPP and EPP (BSATCL = 72.9%, BSACPP = 74.7%,
`
`BSAEPP = 76.2%) does not completely compensate for the loss of the halogen bond observed
`
`for TCL (Table 1 and Figure 4). The relative affinities of the diphenyl ether analogues for
`
`saFabI increase in the order CPP < EPP < TCL. Ki*,app includes the affinity of the more potent
`
`enzyme-inhibitor complex (E-I*) formed following slow isomerization of the initial, rapid-
`
`reversible enzyme-inhibitor complex (E-I; affinity described by Ki
`
`app), according to a two-step
`
`slow-binding mechanism of inhibition. Since saFabI exhibited greatly improved activity and
`
`stability in the presence of glutamate (see Supplemental Discussion), we were able to perform
`
`a more reliable progress curve-based analysis of steady-state inhibition as compared to
`
`previous studies (Heath et al., 2000; Xu et al., 2008).
`
`Reduction of straight-chain substrates by saFabI
`
`Among the straight-chain fatty acyl substrates, we observed an increase in the specificity
`
`constant as the chain length increases from 4 to 8 carbons (Table 3), consistent with previous
`
`results noting a preference for longer chain substrates in the case of S. aureus FabI (Xu et al.,
`
`2008). However, in contrast to other FabI homologues, the specificity constant decreases
`
`beyond a chain length of 8 carbons. These variations are driven by both an increase in kcat and
`
`a decrease in Km, as opposed to ftFabI (Table S3) and ecFabI, where changes in straight-chain
`
`substrate specificity are driven primarily by changes in Km (Rafi et al., 2006; Ward et al.,
`
`1999).
`
`14
`
`Chugai Exhibit 2305
`Pfizer, Inc. v. Chugai Pharmaceutical Co. Ltd.
`IPR2017-01357
`Page 00014
`
`

`

`Substrate specificity of saFabI
`
`The ratio of specificity constants amongst the first-round straight- and branched-chain
`
`substrates of saFabI was approximately 1:24:1 (straight : iso : anteiso) (Table 3). Since the
`
`ratio of velocities vX : vY for competing substrates SX and SY is equal to the ratio of [SX]
`
`kX
`
`cat/KX
`
`m to [SY] kY
`
`cat/KY
`
`m, the available substrate pool also determines the membrane fatty
`
`acid composition (Cornish-Bowden, 2004). FabH, which catalyzes the initial condensation
`
`reaction in the FAS II pathway, controls the pool of substrates that enter the FAS II cycle
`
`(Figure 1). Accordingly, S. aureus and B. subtilis FabH exhibit a preference for branched-
`
`chain acyl-CoA primers over the typical acetyl-CoA primer (Choi et al., 2000; Qiu et al.,
`
`2005). Since the S. aureus membrane is composed mostly of anteiso-C15 fatty acids (Parsons
`
`et al., 2011), our results are consistent with the current thought that FabH is the main
`
`determining factor in branched-chain fatty acid biosynthesis. For the SCFA family of
`
`organisms, FabH tends to prefer the acetyl-CoA primer, resulting in a smaller pool of
`
`available branched-chain substrates (Qiu et al., 2005). Although FabIs from these organisms
`
`are capable of utilizing branched chain (iso-) substrates (Choi et al., 2000; Smirnova and
`
`Reynolds, 2001), the relative rate of BCFA substrate reduction catalyzed by SCFA FabIs like
`
`ftFabI is much lower than for the BCFA family member saFabI (Table S3). Thus, as expected,
`
`the pool of substrates and specificity constants both heavily favor the synthesis and
`
`incorporation of straight-chain fatty acids by this organism.
`
`15
`
`Chugai Exhibit 2305
`Pfizer, Inc. v. Chugai Pharmaceutical Co. Ltd.
`IPR2017-01357
`Page 00015
`
`

`

`Supplemental Discussion
`
`Glutamate sensitivity
`
`We investigated the glutamate sensitivity of saFabI as glutamate is known to be present at
`
`high intracellular concentrations (> 100 mM) in Gram-positive organisms, such as S. aureus,
`
`and has been found to increase the in vitro activity of S. aureus gyrase and topoisomerase IV
`
`(Björklind and Arvidson, 1978; Blanche et al., 1996). Glutamate markedly improved the
`
`catalytic activity of saFabI (~5-fold for 1 M potassium glutamate, ~4-fold for 500 mM
`
`potassium glutamate), such that the maximum kcat obtained was similar to other FabI
`
`homologues with CoA-based substrates (kcat > 500 min-1; Table 3) (Heath et al., 2000; Parikh
`
`et al., 1999; Rafi et al., 2006; Ward et al., 1999; Xu et al., 2008). In contrast, the catalytic
`
`activity of ftFabI is unaffected by glutamate. The reaction velocity of 25 nM ftFabI with 10
`
`µM oct-CoA and 250 µM NADH was 11.25 µM/min both in the presence and absence of 1 M
`
`glutamate. To screen for salt effects, we performed single-point reaction velocity
`
`measurements. Unlike the S. aureus gyrase and topoisomerase IV enzymes (Blanche et al.,
`
`1996), the activation of saFabI by glutamate is not cation-specific though K+ is slightly
`
`preferred over Na+. Among the anions tested, glutamate provided the greatest rate
`
`enhancement. At 50 nM saFabI with 30 µM oct-CoA and 200 µM NADPH, the reaction
`
`velocities relative to sodium glutamate were 0, 0.16, 0.22, 0.49 with sodium chloride, sodium
`
`aspartate, sodium acetate and sodium ADA, respectively. Thus, enhancement of activity is not
`
`a general salt effect but appears to be mediated by a carboxylate motif. Interestingly, there is
`
`evidence of this effect in the literature. To avoid protein precipitation, previous saFabI
`
`purification protocols required buffers containing 100 mM EDTA (Heath et al., 2000; Xu et
`
`al., 2008). Furthermore, some saFabI inhibition assays were performed in the presence of 100
`
`mM sodium ADA pH 6.5 (Payne et al., 2002).
`
`16
`
`Chugai Exhibit 2305
`Pfizer, Inc. v. Chugai Pharmaceutical Co. Ltd.
`IPR2017-01357
`Page 00016
`
`

`

`Notably, there is a physiological link between glutamate and BCFA synthesis - the first step
`
`of BCFA biosynthesis is the transamination of branched-chain amino acids to α-ketoglutarate,
`
`forming the corresponding α-keto acid and glutamate (Figure 1) (Mendoza et al., 2002). We
`
`have shown that glutamate not only enhances enzyme activity but also affects the cofactor
`
`specificity as well as the cooperativity of substrate binding (Figure S5). In fact, the dimer-
`
`tetramer transition may provide a mechanism of regulation by glutamate or similar
`
`metabolites. Based on ambiguous densities in the saFabI P1 crystal structures (Figure S5E)
`
`obtained in the presence of the saFabI-stabilizing anions glutamate, citrate and EDTA which
`
`share the glutaric acid motif, a potential glutamate binding site is located between Arg103 of
`
`SBL-2 and Asn205 of SBL. Molecules occupying this site could interconnect the SBLs,
`
`thereby enhancing substrate binding and stabilizing the tetrameric state. However, future
`
`experiments should clarify the role of glutamate and similar metabolites for saFabI
`
`functioning in vivo.
`
`17
`
`Chugai Exhibit 2305
`Pfizer, Inc. v. Chugai Pharmaceutical Co. Ltd.
`IPR2017-01357
`Page 00017
`
`

`

`Supplemental Experimental Procedures
`
`Cloning
`
`SaFabI from the S. aureus strain NCTC 8325 was purified using the safabINCTC8325:pET-
`
`16b(+) plasmid (Xu et al., 2008). We also amplified the same safabI gene from S. aureus
`
`strain N315 via PCR (originating from safabIN315:pET-16b(+)) and cloned it into pETM-11
`(EMBL) using the NcoI and HindIII (New England Biolabs) restriction sites (primers: 5‘-
`CGAAGGTCGTCCCATGGTAAATCTTGAAAAC-3‘
`5'-
`and
`CCAATAACGTGAACAAAGCTTCTGAATG-3' (Biomers)). The saFabI isoform from S. aureus
`
`NCTC 8325 was used only for kinetic experiments. It differs from the S. aureus N315 isoform
`
`by a single amino acid (G191S). Although residue 191 is positioned in close proximity to the
`
`nicotinamide ring of NADPH, the Km of saFabI-G191 with respect to NADPH was identical
`
`to that reported previously under similar assay conditions for saFabI-S191 (Xu et al., 2008).
`
`Expression and purification
`
`E. coli BL21 (DE3) bacteria (Novagen) containing the safabI:pETM-11 plasmid were grown
`
`in Luria-Bertani (LB) medium (Lennox) supplemented with 50 µg/ml kanamycin at 37 °C.
`
`After induction with 0.5 mM isopropyl-β-D-1-thiogalactopyranoside (IPTG) at an OD600 nm of
`
`0.6 the temperature was reduced to 18 °C. Cells were harvested after 18 hours post-induction,
`
`resuspended in lysis buffer (50 mM Tris-HCl pH 8.0, 500 mM NaCl) and lysed through a cell
`
`disruptor at 1.5 kbar. The lysate was centrifuged at 50,000 g and the supernatant loaded onto a
`
`Protino Ni-TED column (Macherey-Nagel). After washing (50 mM Tris-HCl pH 8.0, 1 M
`
`NaCl), the protein was eluted (50 mM Tris-HCl pH 8.0, 500 mM NaCl, 250 mM imidazole).
`
`A buffer exchange with PD-10 desalting columns (Amersham Biosciences) was followed by
`
`size exclusion chromatography (SEC) on a HiLoad 26/60 Superdex 200 pg column (GE
`
`Healthcare) pre-equilibrated with storage buffer (20 mM trisodium citrate pH 5.6, 280 mM
`
`18
`
`Chugai Exhibit 2305
`Pfizer, Inc. v. Chugai Pharmaceutical Co. Ltd.
`IPR2017-01357
`Page 00018
`
`

`

`NaCl, 1mM EDTA, 280 mM potassium glutamate). Protein containing fractions were pooled
`
`and concentrated to 15 mg/ml prior to shock-freezing in liquid nitrogen and storage at -80 °C.
`
`For further details see also Table S4 (this protocol refers to procedure D in Table S4).
`
`Crystallization, data collection and structure determination
`
`In a vapor diffusion experiment containing equal amounts of protein and precipitant solution
`
`(0.1 M K/Na-phosphate pH 6.5, 35% 2-methyl-2,4-pentanediol (MPD)), a single TCL-1
`
`crystal (nomenclature according to Table S2) of space group P212121 grew within 3 weeks.
`
`Diffraction data of the directly flash-frozen crystal were collected to 2.8 Å at the MX
`
`beamline 14.1 at BESSY II (Berlin) utilizing a MarMosaic 225 detector (λ = 0.976 Å, T = 100
`
`K). Data were processed with iMosflm (Leslie, 1992) and Scala (Evans, 2006). Phases were
`
`determined with Phaser (McCoy et al., 2007) by molecular replacement using the FabI
`
`structure of B. anthracis (PDB code: 2QIO:A) as search model (Tipparaju et al., 2008).
`
`Alternate cycles of maximum likelihood refinement and model building were performed in
`
`Refmac 5 (Murshudov et al., 1997) and Coot (Table S2) (Emsley and Cowtan, 2004).
`
`Refinement was improved with the implementation of TLS-refinement and NCS-restraints
`
`excluding Met99. After the first rounds of refinement triclosan, NADP+ and water molecules
`
`were included into the model according to the Fo-Fc map.
`
`Rod-shaped apo-1 crystals of space group P32 were obtained in a vapor diffusion setup using
`
`0.61 M NH4H2PO4, 9% t-butanol, 0.01 M EDTA pH 7.0 as precipitant. For data collection at
`
`ID 29 (λ = 0.919 Å, T = 100 K) of the European Synchrotron Radiation Facility (ESRF,
`
`Grenoble) the crystals were briefly transferred into cryoprotectant solution (0.61 M
`
`NH4H2PO4, 9% t-butanol, 0.01 M EDTA pH 7.0, 25% glycerol) and cryocooled in liquid
`
`nitrogen. After data processing with XDS (Kabsch, 1993), twinning was detected using
`
`Phenix.xtriage (Zwart et al., 2008). Phaser molecular replacement was performed with the
`
`TCL-1 structure omitting regions 95-108, 149-158 and 190-221 of the protein as well as all
`
`19
`
`Chugai Exhibit 2305
`Pfizer, Inc. v. Chugai Pharmaceutical Co. Ltd.
`IPR2017-01357
`Page 00019
`
`

`

`small molecules. To avoid model bias due to high non-crystallographic symmetry (12
`
`molecules per asymmetric unit) and twinning, Rfree flags were assigned in 20 thin resolution
`
`shells. Model building in Coot was alternated with a combined refinement using Phenix.refine
`
`(Adams et al., 2010) and Refmac 5 (Table S1). As no additional electron density for the
`
`cofactor and inhibitor was observed, this structure is referred to as the apo-form.
`
`Cube-shaped apo-2 crystals of space group P43212 grew in 1.15 M (NH4)2SO4, 0.1 M sodium
`
`acetate pH 5.0, 1% ethanol as precipitant in a vapor diffusion experiment. Crystals were
`
`cryocooled (30% glycerol) and diffraction data were collected to 2.45 Å at beamline ID 29
`
`(ESRF) using an ADSC Quantum Q315r detector (λ = 0.919 Å, T = 100 K). Data were
`
`analyzed as described for the apo-1 structure and refined (including TLS refinement) with
`
`Phenix.refine (Table S1). As in the case of apo-1, no interpretable cofactor or inhibitor density
`
`was present.
`
`Ternary saFabI-NADP+-EPP crystals were obtained with 0.1 M K/Na-phosphate pH 6.5 and
`
`36% MPD as precipitant. Data were collected with an ADSC Quantum Q315r detector at
`
`beamline ID 14-4 (ESRF, λ = 0.980 Å, T = 100 K) and were processed to 1.9 Å using
`
`iMosflm and Scala. Molecular replacement was performed with Phaser using the TCL-1
`
`structure lacking all small molecules as search model. Refinement (including TLS refinement
`
`(Painter and Merritt, 2006)) and model building were performed using Refmac 5 and Coot
`
`(Table S2). NADP+ and EPP were included in the model (Schuttelkopf and van Aalten, 2004)
`
`after molecular replacement and initial refinement. Additionally, the Fo-Fc map indicated an
`
`alternative conformation of the amino acid stretch 99-105. The second conformation was
`
`modeled for all subunits with clear densities for this feature.
`
`A vapor diffusion experiment with TCL-treated saFabI protein and a similar precipitant
`
`solution (0.1 M K/Na-phosphate pH 6.5 and 38% MPD) resulted in TCL-2 crystals of space
`
`group P1. Diffraction data were collected to 2.1 Å at beamline 14.1 (BESSY II) using a
`
`MarMosaic 225 detector (λ = 0.918 Å, T = 100 K) and processed with iMosflm and Scala.
`
`20
`
`Chugai Exhibit 2305
`Pfizer, Inc. v. Chugai Pharmaceutical Co. Ltd.
`IPR2017-01357
`Page 00020
`
`

`

`The EPP structure without alternative conformations served as a search model for molecular
`
`replacement in Phaser. Refmac 5 and Coot were used for refinement (including TLS
`
`refinement) and model building (Table S2).
`
`Similarly, CPP crystals of saFabI were grown with a precipitant solution containing 0.1 M
`
`K/Na-phosphate pH 6.5 and 35% MPD. Diffraction data of a directly cryocooled crystal were
`
`collected at beamline ID 29 (ESRF) with a PILATUS 6M detector (λ = 0.976 Å, T = 100 K).
`
`Indexing and integration were performed with the XDS package. Scala was used for scaling.
`
`Structure determination and refinement followed the procedure as described for TCL-2 (Table
`
`S2).
`
`As saFabI crystallized in space group P1 with similar cell parameters for the different
`
`inhibitors, comparability is warranted (Table S2). For further analysis we used the subunits H
`
`of the different structures which display no significant crystal contacts at the active site and
`
`therefore approximate the natural state in solution despite the increased flexibility that results
`
`in weaker electron densities. Distances and angles were measured for all subunits and are
`
`given as mean values ± standard deviations.
`
`To avoid model bias, omit maps were calculated directly after molecular replacement and an
`
`initial refinement prior to inclusion of any small molecule (Figure S2). All structural figures
`
`were prepared with PyMol (DeLano, 2002).
`
`Steady-state kinetic assays
`
`Kinetic parameters for the wild-type saFabI catalyzed reduction of trans-2-octenoyl-CoA
`
`(oct-CoA) using NADPH were determined by varying the concentration of oct-CoA at several
`
`fixed concentrations of NADPH. Previous mechanistic studies suggest that saFabI catalyzes
`
`its reaction via an ordered Bi Bi mechanism with the fatty acyl substrate binding first (Xu et
`
`al., 2008). However, our structural studies suggest that NADP+ likely binds first, followed by
`
`the inhibitor. We believe this extends to substrate binding with NADPH binding before the
`
`21
`
`Chugai Exhibit 2305
`Pfizer, Inc. v. Chugai Pharmaceutical Co. Ltd.
`IPR2017-01357
`Page 00021
`
`

`

`fatty acyl substrate, in accordance with other FabI homologues. Thus, the data were fitted to
`
`equations 2 and 3, assuming an ordered B

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket